WO2012125613A1 - Morpholine-spirocyclic piperidine amides as modulators of ion channels - Google Patents

Morpholine-spirocyclic piperidine amides as modulators of ion channels Download PDF

Info

Publication number
WO2012125613A1
WO2012125613A1 PCT/US2012/028882 US2012028882W WO2012125613A1 WO 2012125613 A1 WO2012125613 A1 WO 2012125613A1 US 2012028882 W US2012028882 W US 2012028882W WO 2012125613 A1 WO2012125613 A1 WO 2012125613A1
Authority
WO
WIPO (PCT)
Prior art keywords
pain
compound
och
mmol
alkyl
Prior art date
Application number
PCT/US2012/028882
Other languages
English (en)
French (fr)
Inventor
Sara Sabina Hadida-Ruah
Hayley Marie Binch
Michael Paul Deninno
Lev Tyler Dewey Fanning
Bryan A. Frieman
Peter Diederik Jan Grootenhuis
Nicole Hilgraf
Pramod Joshi
Edward Adam Kallel
Joseph Pontillo
Alina Silina
Urvi Jagdishbhai Sheth
Dennis James Hurley
Vijayalaksmi Arumugam
Mark Thomas Miller
Original Assignee
Vertex Pharmaceuticals Incorporated
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to MX2013010476A priority Critical patent/MX347982B/es
Priority to NZ615014A priority patent/NZ615014B2/en
Priority to JP2013558104A priority patent/JP6088443B2/ja
Priority to CN201280018832.5A priority patent/CN103517910B/zh
Priority to ES12710841.3T priority patent/ES2618929T3/es
Priority to CA2829803A priority patent/CA2829803A1/en
Priority to AU2012229187A priority patent/AU2012229187B2/en
Priority to EP12710841.3A priority patent/EP2686325B1/en
Publication of WO2012125613A1 publication Critical patent/WO2012125613A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the invention relates to compounds useful as inhibitors of ion channels.
  • the invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.
  • Pain is a protective mechanism that allows healthy animals to avoid tissue damage and to prevent further damage to injured tissue. Nonetheless there are many conditions where pain persists beyond its usefulness, or where patients would benefit from inhibition of pain. Voltage-gated sodium channels are believed to play a critical role in pain signaling. This belief is based on the known roles of these channels in normal physiology, pathological states arising from mutations in sodium channel genes, preclinical work in animal models of disease, and the clinical usefulness of known sodium channel modulating agents (Cummins, T. R., Sheets, P. L., and Waxman, S. G., The roles of sodium channels in nociception: Implications for mechanisms of pain.
  • NaV's Voltage-gated sodium channels
  • NaV's are key biological mediators of electrical signaling.
  • NaV's are the primary mediators of the rapid upstroke of the action potential of many excitable cell types (e.g. neurons, skeletal myocytes, cardiac myocytes), and thus are critical for the initiation of signaling in those cells (Hille, Bertil, Ion Channels of Excitable Membranes, Third ed. (Sinauer Associates, Inc., Sunderland, MA, 2001)).
  • antagonists that reduce NaV currents can prevent or reduce neural signaling.
  • NaV channels are considered likely targets in pathologic states where reduced excitability is predicted to alleviate the clinical symptoms, such as pain, epilepsy, and some cardiac arrhythmias (Chahine, M., Chatelier, A., Babich, O., and Krupp, J. J., Voltage-gated sodium channels in neurological disorders. CNS Neurol Disord Drug Targets 7 (2), 144 (2008)).
  • the NaV's form a subfamily of the voltage-gated ion channel super- family and comprises 9 isoforms, designated NaV 1.1 - NaV 1.9. The tissue localizations of the nine isoforms vary greatly.
  • NaV 1.4 is the primary sodium channel of skeletal muscle
  • NaV 1.5 is primary sodium channel of cardiac myocytes.
  • NaV's 1.7, 1.8 and 1.9 are primarily localized to the peripheral nervous system, while NaV's 1.1 , 1.2, 1.3, and 1.6 are neuronal channels found in both the central and peripheral nervous systems.
  • the functional behaviors of the nine isoforms are similar but distinct in the specifics of their voltage-dependent and kinetic behavior (Catterall, W. A., Goldin, A. L., and Waxman, S. G., International Union of Pharmacology. XLVII.
  • NaV channels have been identified as the primary target for some clinically useful pharmaceutical agents that reduce pain (Cummins, T. R., Sheets, P. L., and Waxman, S. G., The roles of sodium channels in nociception: Implications for mechanisms of pain. Pain 131 (3), 243 (2007)).
  • the local anesthetic drugs such as lidocaine block pain by inhibiting NaV channels.
  • These compounds provide excellent local pain reduction but suffer the drawback of abolishing normal acute pain and sensory inputs.
  • Systemic administration of these compounds results in dose limiting side effects that are generally ascribed to block of neural channels in the CNS (nausea, sedation, confusion, ataxia). Cardiac side effects can also occur, and indeed these compounds are also used as class 1 anti-arrhythmics, presumably due to block of
  • NaVl .5 channels in the heart Other compounds that have proven effective at reducing pain have also been suggested to act by sodium channel blockade including
  • Anticonvulsants aspects of their mechanisms of action. Eur J Pain 6 Suppl A, 3 (2002); Wang, G. K., Mitchell, J., and Wang, S. Y., Block of persistent late Na + currents by antidepressant sertraline and paroxetine. J Membr Biol 222 (2), 79 (2008)). These compounds are likewise dose limited by adverse effects similar to those seen with the local anesthetics. Antagonists that specifically block only the isoform(s) critical for nocioception are expected to have increased efficacy since the reduction of adverse effects caused by block of off-target channels should enable higher dosing and thus more complete block of target channels isoforms.
  • NaV 1.3 is normally found in the pain sensing neurons of the dorsal root ganglia (DRG) only early in development and is lost soon after birth both in humans and in rodents. Nonetheless, nerve damaging injuries have been found to result in a return of the NaV 1.3 channels to DRG neurons and this may contribute to the abnormal pain signaling in various chronic pain conditions resulting from nerve damage (neuropathic pain). These data have led to the suggestion that pharmaceutical block of NaV 1.3 could be an effective treatment for neuropathic pain.
  • DRG dorsal root ganglia
  • NaV 1.7, 1.8, and 1.9 are highly expressed in DRG neurons, including the neurons whose axons make up the C-fibers and ⁇ nerve fibers that are believed to carry most pain signals from the nocioceptive terminals to the central nervous. Like NaV 1.3, NaV 1.7 expression increases after nerve injury and may contribute to neuropathic pain states. The localization of NaV 1.7, 1.8, and 1.9 in nocioceptors led to the hypothesis that reducing the sodium currents through these channels might alleviate pain. Indeed, specific interventions that reduce the levels of these channels have proven effective in animal models of pain.
  • NaV 1.9 global knock out mice have decreased sensitivity to
  • NaV 1.1 and NaV 1.2 mutations result in various forms of epilepsy (Fujiwara, T., Clinical spectrum of mutations in SCN1 A gene: severe myoclonic epilepsy in infancy and related epilepsies. Epilepsy Res 70 Suppl 1, S223 (2006); George, A. L., Jr., Inherited disorders of voltage-gated sodium channels. J Clin Invest 115 (8), 1990 (2005); Misra, S. N., Kahlig, . M., and George, A.
  • Brugada Syndrome and long QT syndrome (Bennett, P. B., Yazawa, K., Makita, N., and George, A. L., Jr., Molecular mechanism for an inherited cardiac arrhythmia. Nature 376 (6542), 683 (1995); Darbar, D. et al, Cardiac sodium channel (SCN5A) variants associated with atrial fibrillation. Circulation 111 (15), 1927 (2008); Wang, Q. et al., SCN5A mutations associated with an inherited cardiac arrhythmia, long QT syndrome. Cell 80 (5), 805 (1995)).
  • NaV 1.7 null mutations in human patients were recently described by several groups (Ahmad, S. et al., A stop codon mutation in SCN9A causes lack of pain sensation. Hum Mol Genet 16 (17), 2114 (2007); Cox, J. J. et al., An SCN9A channel opathy causes congenital inability to experience pain. Nature 444 (7121), 894 (2006); Goldberg, Y. P. et al., Loss-of-function mutations in the NaV 1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet 71 (4), 311 (2007)). In all cases patients exhibit congenital indifference to pain. These patients report no pain under any circumstances.
  • These compounds and pharmaceutically acceptable compositions are useful for treating or lessening the severity of a variety of diseases, disorders, or conditions, including, but not limited to, acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, or cancer pain.
  • diseases, disorders, or conditions including, but not limited to, acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trige
  • the invention provides compounds of formula I:
  • R 1 is C1-C6 alkyl, C1-C6 fluoroalkyl, C3-C8 cycloalkyl, halo, CN, NR 8 S0 2 R 8 ,
  • R 2 is H, C1-C6 alkyl, C1-C6 fluoroalkyl, CF 3 , optionally substituted cycloalkyl, aryl, heteroaryl or heterocycloalkyl, COR 8 , C0 2 R 8 , CON(R 8 ) 2 , CF 3 , CHF 2 , or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ; R 3 is C1-C6 alkyl or halo;
  • R 8 is H, C1-C6 alkyl, or C3-C8 cycloalkyl, a straight chain, branched, or cyclic (Cl- C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR, or 2 R 8 taken together with the atoms to which they are attached form a ring;
  • R 9 is H, CF 3 , CHF 2 , CH 2 F, C0 2 R, OH, optionally substituted aryl, heteroaryl, C3-C8 cycloalkyl, heterocycloalkyl, N(R) 2 , NRCOR, CON(R) 2 , CN, or S0 2 R;
  • R is H, C1-C6 alkyl, optionally substituted aryl, heteroaryl, C3-C8 cycloalkyl, or heterocycloalkyl;
  • A is an optionally substituted aryl, heteroaryl or heterocyclic; n is an integer from 0 to 4 inclusive; and o is an integer from 0 to 4 inclusive.
  • R 1 is C1-C6 alkyl, C1-C6 fluoroalkyl, C3-C8 cycloalkyl, CN, CON(R 8 ) 2 , S0 2 N(R 8 ) 2 , CF 3 , optionally substituted heterocycloalkyl, phenyl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 , or two R 1 taken together form an oxo group.
  • compounds of the invention can optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention.
  • substituents such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention.
  • the phrase "optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.”
  • the variables R ⁇ R 9 in formula I encompass specific groups, such as, for example, alkyl and aryl. Unless otherwise noted, each of the specific groups for the variables R'-R 8 can be optionally substituted with one or more substituents of halo, cyano, oxoalkoxy, hydroxy, amino, nitro, aryl, haloalkyl, and alkyl.
  • an alkyl group can be optionally substituted with one or more of halo, cyano, oxoalkoxy, hydroxy, amino, nitro, aryl, haloalkyl, and alkyl.
  • an aryl group can be optionally substituted with one or more of halo, cyano, alkoxy, hydroxy, nitro, haloalkyl, and alkyl.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and preferably their recovery, purification, and use for one or more of the purposes disclosed herein.
  • a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • the two alkoxy groups can form a ring together with the atom(s) to which they are bound.
  • an optionally substituted group can have a substituent at each substitutable position of the group, and when more than one position in any given structure can be substituted with more than one substituent selected from a specified group, the substituent can be either the same or different at every position.
  • a ring substituent such as a heterocycloalkyl, can be bound to another ring, such as a cycloalkyl, to form a spiro-bicyclic ring system, e.g., both rings share one common atom.
  • substituents envisioned by this invention are those combinations that result in the formation of stable or chemically feasible compounds.
  • aliphatic refers to zero or any integer number that is equal or less than the number following the phrase.
  • up to 3 means any one of 0, 1 , 2, and 3.
  • aliphatic means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation.
  • aliphatic groups contain 1-20 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-10 aliphatic carbon atoms.
  • aliphatic groups contain 1 -8 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-6 aliphatic carbon atoms, and in yet other embodi ments aliphatic groups contain 1-4 aliphatic carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups. The term "cycloaliphatic" or
  • cycloalkyl mean a monocyclic hydrocarbon, bicyclic, or tricyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic and has a single point of attachment to the rest of the molecule.
  • cycloaliphatic refers to a monocyclic C3-C8 hydrocarbon or bicyclic C 8 -C 12 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule wherein any individual ring in said bicyclic ring system has 3-7 members.
  • electronegative group means an atom or a group that is electronegative relative to hydrogen. See, e.g., “Advanced Organic Chemistry: Reactions, Mechanisms, and Structure," Jerry March, 4 th Ed., John Wiley & Sons (1 92), e.g., pp. 14-16, 18-19, etc. Exemplary such substituents include halo such as CI, Br, or F, CN, COOH, CF 3 , etc.
  • heterocycle means non- aromatic, monocyclic, bicyclic, or tricyclic ring systems in which one or more ring atoms in one or more ring members is an independently selected heteroatom.
  • Heterocyclic ring can be saturated or can contain one or more unsaturated bonds.
  • heterocycloalkyl'Or “heterocyclic” group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the ring system contains 3 to 7 ring members.
  • heteroatom means oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
  • alkoxy refers to an alkyl group, as previously defined, attached to the principal carbon chain through an oxygen (“alkoxy”) or sulfur (“thioalkyl”) atom.
  • alkoxy refers to an alkyl group, as previously defined, attached to the principal carbon chain through an oxygen (“alkoxy”) or sulfur (“thioalkyl”) atom.
  • aryl used alone or as part of a larger moiety as in “aralkyl”,
  • aralkoxy refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring carbon atoms, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring carbon atoms.
  • aryl may be used interchangeably with the term “aryl ring”.
  • heteroaryl used alone or as part of a larger moiety as in “heteroaralkyl” or
  • heteroarylalkoxy refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, and wherein each ring in the system contains 3 to 7 ring members.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or the term “heteroaromatic”.
  • alkylidene chain refers to a straight or branched carbon chain that may be fully saturated or have one or more units of unsaturation and has two points of attachment to the rest of the molecule.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds of formula I wherein one or more hydrogen atoms are replaced deuterium or tritium, or one or more carbon atoms are replaced by a 13 C- or I4 C-enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools, probes in biological assays, or sodium channel blockers with improved therapeutic profile.
  • R group can be bonded to any carbon, or if applicable, heteroatom such as N, of that ring as valency allows.
  • R 1 , R 2 , R 3 , R 4 , R 5 , or R 6 when a CH 2 unit or, interchangeably, methylene unit may be replaced by O, CO, S, SO, S0 2 or NR 8 , it is meant to include any CH 2 unit, including a CH 2 within a terminal methyl group.
  • -CH 2 CH 2 CH 2 SH is within the definition of C1 -C6 alkyl wherein up to two CH 2 units may be replaced by S because the CH 2 unit of the terminal methyl group has been replaced by S.
  • the invention features a compound of formula ⁇ and the attendant definitions, wherein R 1 is halo or optionally substituted aryl, heteroaryl, C1-C6 alkyl, C1-C6 fluoroalkyl, a straight chain, branched, or cyclic (Cl- C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, or NR , or 2 R taken together form an oxo group.
  • R is F or optionally substituted phenyl, pyridyl, oxazole, thiazole, pyrazole, oxadiazole,
  • the invention features a compound of formula I and the attendant definitions, wherein R 2 is H, C1-C6 alkyl, C1-C6 fluoroalkyl, CF 3 , an optionally substituted cycloalkyl, aryl, heterocycloalkyl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, or NR 8 .
  • R 2 is H, CH 2 CHF 2 , CH 2 CF 3 , CH(CH 3 )CH 2 F, CH 2 CH(CH 3 ) 2 , CH 3 , CH 2 CH 3 , tBu, CH 2 CN, CH(CH 3 ) 2 ,
  • CH 2 CH 2 OCH 3 CH 2 OCH 3 , CH 2 C(0)CH 3 , CH 2 C(0)0CH 3 , CH 2 CH 2 OCH 2 CH 2 CH 3 , CH CCCH 2 CH 3 , CH 2 CH 2 OCH 2 CH 3 , CH 2 CH 2 SCH 3 , CH 2 CH 2 CH 2 OCH 3 ,
  • the invention features a compound of formula I and the attendant definitions, wherein n is 0, 1 , 2, or 3. In another embodiment, n is 1 or 2. In another embodiment, n is 1. [0040] In another embodiment, the invention features a compound of formula I and the attendant definitions, wherein o is 0 or 1. In another embodiment, o is 0.
  • the invention features a compound of formula I and the attendant definitions, wherein A is
  • R 4 is H, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 NR 8 S0 2 R 8 , S0 2 R 8 , SOR 8 , SR 8 , C0 2 R 8 , NR 8 COR 8 , NR 8 C0 2 R 8 , C0N(R 8 ) 2 , S0 2 N(R 8 ) 2 , CHF 2 , CF 3 , OCF 3 , OCHF 2 , R 9 , heterocycloalkyl,
  • R 5 is H, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , NR 8 S0 2 R 8 , S0 2 R 8 , SOR 8 , SR 8 , C0 2 R 8 , NR 8 COR 8 ,
  • NR 8 C0 2 R 8 CON(R 8 ) 2 , S0 2 N(R 8 ) 2 , CF 3 , OCF 3 , OCHF 2 , R 9 , heterocycloalkyl, heterocycloalkoxy, aryl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 6 is H, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , NR 8 S0 2 R 8 , S0 2 R 8 , SOR 8 , SR 8 , C0 2 R 8 , NR 8 COR 8 , NR 8 C0 2 R 8 , CON(R 8 ) 2 , S0 2 N(R 8 ) 2 , CF 3 , OCF 3 , OCHF 2 , R 9 , heterocycloalkyl, heterocycloalkoxy, aryl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ; or two occurrences of R 4 and R 5 , or R 5 and R 6 together
  • R 4 is H, C1-C6 alkyl, C1 -C6 alkoxy, halo, OCF 3 , OCHF 2 , or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, or NR 7 .
  • Cl-C8-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, or NR 7 .
  • R 4 is H, F, CH 3 , OCH 3 , OCF 3 , or OCHF 2 .
  • R 5 is H, C1-C6 alkyl, C1 -C6 alkoxy, halo, CF 3 , CN, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 , N, or NR 8 .
  • R 5 is H, CH 3 , OCH 3 , OCH 2 CH 3 , OCH(CH 3 ) 2 , F, CI, CN, CF 3 , or CH 2 OH.
  • R 6 is H, C1-C6 alkyl, C1-C6 alkoxy, S0 2 R 8 , S0 2 N(R 8 ) 2 , R 9 , or a straight chain, branched, or cyclic (Cl-C8)-R 9 , wherein up to three CH 2 units may be replaced with O, S, SO, S0 2 , N, or NR 8 .
  • R 6 is H, CH 2 OH, OCH 3 , OCH 2 CH 3 , OCH 2 CH 2 CH 3 , 0CH 2 CH 2 CH(CH 3 ) 2 , OtBu, tBu, OCH(CH 3 ) 2 , OCH 2 C(CH 3 ) 2 OCH 3 , CH(OH)CH(CH 3 ) 2 , C(OH)(CH 2 CH 3 ) 2 ,
  • the invention features a compound of formula I
  • the invention features a compound of formula I and the attendant definitions, wherein A is heteroaryl or heterocyclic.
  • A is a monocyclic heteroaryl comprising 1 to 3 heteroatoms
  • A is selected from a bicyclic heteroaryl comprising from 1 to 3 heteoratoms independently selected from N, O, or S.
  • A is wherein:
  • R 4 is H, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , NR 8 S0 2 R 8 , S0 2 R 8 , SOR 8 , SR 8 , C0 2 R 8 , NR 8 COR 8 , NR 8 C0 2 R 8 , CON(R 8 ) 2 , S0 2 N(R 8 ) 2 , CHF 2 , CF 3 , OCF 3 , OCHF 2 , R 9 , heterocycloalkyl,
  • heterocycloalkoxy aryl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 5 is H, C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, C3-C8 cycloalkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , NR 8 S0 2 R 8 , S0 2 R 8 , SOR 8 , SR 8 , C0 2 R 8 , NR 8 COR 8 ,
  • NR 8 C0 2 R 8 CON(R 8 ) 2 , S0 2 N(R 8 ) 2 , CF 3 , OCF 3 , OCHF 2 , R 9 , heterocycloalkyl, heterocycloalkoxy, aryl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 6 is H, CI -C6 alkyl, C3-C8 cycloalkyl, CI -C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , NR 8 S0 2 R 8 , S0 2 R 8 , SOR 8 , SR 8 , C0 2 R 8 , NR 8 COR 8 , NR 8 C0 2 R 8 , CON(R 8 ) 2
  • R 4 is H or C1-C6 alkyl.
  • R 4 is H.
  • R 5 is H, C1-C6 alkyl, or C1-C6 alkoxy. In another embodiment, R 5 is H, CH 3 , or OCH 3 .
  • R 6 is H, CN, C1-C6 alkoxy, or CF 3 . In another embodiment, R 6 is H, CN, OCH 3 , or CF 3 . [0051] In another embodiment, the invention features a compound of formula I and the attendant definitions, wherein A is selected from the following:
  • the invention features the compounds of formula I and the attendant definitions, wherein the compound has formula IA:
  • R is H, C1-C6 alkyl, C1-C6 fluoroalkyl, an optionally substituted cycloalkyl, aryl, heteroaryl, or heterocycloalkyl, or a straight chain, branched, or cyclic (Cl- C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 5 is H, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 fluoroalkyl, halo, CF 3 , OCF 3 , OCHF 2 , or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 6 is H, C1-C6 alkyl, C1-C6 alkoxy, CN, S0 2 R 8 , CON(R 8 ) 2 , S0 2 N(R 8 ) 2 ,
  • heterocycloalkyl or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR. 8 ;
  • R 7 is C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , CF 3 , OCF 3 , or OCHF 2 ; and p is an integer from 0 to 3 inclusive.
  • R 2 is selected from C1-C6 alkyl, C1-C6 fluoroalkyl, cycloalkyl, aryl, heterocycloalkyl, heteroaryl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 2 is CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , CH 2 CH(CH 3 ) 2 , CH 2 CHF 2 , CH 2 CF 3 , CH(CH 3 )CH 2 F, CH 2 CN, CH 2 CH 2 OH,
  • R 5 is selected from H, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 fluroroalkyl, halo, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 5 is H, CH 3 , OCH 3 , OCH 2 CH 3 , CF 3 , CI, F, or CH 2 OH.
  • R 6 is selected from H, C1-C6 alkoxy, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 6 is H, CH 2 OH, OCH 2 CH 3 , OtBu, OCH(CH 3 ) 2 , OCH 2 C(CH 3 ) 2 OCH 3 , CH(OH)CH(CH 3 ) 2 , OCH 2 C(CH 3 ) 2 OH, C(CH 3 ) 2 OH, OCH 2 CH 2 OCH 3 , OCH 2 CH 2 OH, OCH 2 CH 2 CH 2 OH, CCCH OCH 3 , S0 2 CH 3 , S0 2 CH 2 CH(CH 3 ) 2 , S0 2 CH(CH 3 ) 2 , S0 2 CH 2 CH 3 , S0 2 C(CH 3 ) 3
  • R 7 is halo. In another embodiment, R 7 is F.
  • the moiety is selected from:
  • the invention features a compound of formula I and the attendant definitions of the above embodiments, wherein the compound has formula IB:
  • R is H, C1-C6 alkyl, C1-C6 fluoroalkyl, an optionally substituted cycloalkyl, aryl, heteroaryl, or heterocycloalkyl, or a straight chain, branched, or cyclic (Cl- C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 5 is H, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 fluoroalkyl, halo, CF 3 , OCF 3 , OCHF 2 , or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ; R 6 is H, C1-C6 alkyl, C1-C6 alkoxy, CN, S0 2 R 8 , CON(R 8 ) 2 , S0 2 N(R 8 ) 2 ,
  • heterocycloalkyl or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 7 is C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , CF 3 , OCF 3 , or OCHF 2 ; and
  • p is an integer from 0 to 3 inclusive.
  • R 2 is selected from C1-C6 alkyl, C1-C6 fluoroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, or a straight chain, branched, or cyclic (CI -C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 2 is CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , CH 2 CH(CH 3 ) 2 , CH 2 CHF 2 , CH 2 CF 3 , CH(CH 3 )CH 2 F, CH 2 CN, CH 2 CH 2 OH,
  • R is selected from H, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 fluroroalkyl, halo, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 5 is H, CH 3 , OCH 3 , OCH 2 CH 3 , CF 3 , CI, F, or CH 2 OH.
  • R 6 is selected from H, C1-C6 alkoxy, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 6 is H, CH 2 OH, OCH 2 CH 3 , OtBu, OCH(CH 3 ) 2 , OCH 2 C(CH 3 ) 2 OCH 3 , CH(OH)CH(CH 3 ) 2 , OCH 2 C(CH 3 ) 2 OH, C(CH 3 ) 2 OH, OCH 2 CH 2 OCH 3 , OCH 2 CH 2 OH, OCH 2 CH 2 CH 2 OH, CCCH OCH 3 , S0 2 CH 3 , S0 2 CH 2 CH(CH 3 ) 2 , S0 2 CH(CH 3 ) 2 , S0 2 CH 2 CH 3 , S0 2 C(CH 3 ) 3
  • R is halo. In another embodiment, R is F.
  • the Het ring is a mono or bi cyclic optionally substituted heterocyclic or heteroaryl ring
  • R 2 is H, C1-C6 alkyl, C1-C6 fluoroalkyl, an optionally substituted aryl, heteroaryl, or heterocycloalkyl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 5 is H, C1-C6 alkyl, C1-C6 alkoxy, halo, CF 3 , OCF 3 , OCHF 2 , or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 6 is H, C1-C6 alkyl, C1-C6 alkoxy, CN, S0 2 R 8 , CON(R 8 ) 2 , S0 2 N(R 8 ) 2 ,
  • heterocycloalkyl or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8
  • R 7 is C1 -C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , CF 3 , OCF 3 , or OCHF 2 ; and p is an integer from 0 to 3 inclusive.
  • the Het ring is an optionally substituted thiazole, pyridine, pyrazole, oxazole, or oxadiazole.
  • p is 0 or 1.
  • R 7 is C1-C6 alkyl.
  • R 7 is CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , or tBu.
  • the Het ring is
  • R 2 is selected from C1-C6 alkyl or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O CO S SO 0 CF or NR 8 .
  • R 2 is CH 2 CH 3 , tBu,
  • R 5 is selected from H, C1-C6 alkyl, C1-C6 alkoxy, or halo. In another embodiment, R 5 is H, CH 3 , OCH 3 , F, or CI.
  • R 6 is H, C1-C6 alkoxy, or a straight chain, branched, or cyclic (CI -C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 .
  • R 6 is OCH(CH 3 ) 2 ,
  • the invention features a compound of formula I and the attendant definitions, wherein the compound has formula ID:
  • Het ring is a mono or bicyclic optionally substituted heterocyclic or heteroaryl ring
  • R 2 is H, C1-C6 alkyl, C1-C6 fluoroalkyl, an optionally substituted aryl, heteroaryl, or heterocycloalkyl, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to two CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 5 is H, C1-C6 alkyl, C1-C6 alkoxy, halo, CF 3 , OCF 3 , OCHF 2 , or a straight chain, branched, or cyclic (Cl -C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8 ;
  • R 6 is H, C1-C6 alkyl, C1 -C6 alkoxy, CN, S0 2 R 8 , CON(R 8 ) 2 , S0 2 N(R 8 ) 2 ,
  • heterocycloalkyl or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF 2 , or NR 8
  • R 7 is C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 alkoxy, halo, CN, OH, OR 8 , N(R 8 ) 2 , CF 3 , OCF 3 , or OCHF 2 ; and p is an integer from 0 to 3 inclusive.
  • the Het ring is an optionally substituted thiazole, pyridine, pyrazole, oxazole, or oxadiazole.
  • p is 0 or 1 ,
  • R 7 is C1-C6 alkyl. [0077] In another embodiment, R 7 is CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , or tBu.
  • the Het ring is
  • R 2 is CH 2 CH 3 , tBu,
  • R 5 is selected from H, C1-C6 alkyl, C1-C6 alkoxy, or halo.
  • R 5 is H, CH 3 , OCH 3 , F, or CI.
  • R 6 is H, C1-C6 alkoxy, or a straight chain, branched, or cyclic (Cl-C8)-R 9 wherein up to three CH 2 units may be replaced with O, CO, S, SO, S0 2 , N, CF or NR 8 .
  • R 6 is OCH(CH 3 ) 2 , C(CH 3 ) 2 OH, OCH 2 CH 2 OH,
  • the invention features a compound of formula I and the attendant definitions, wherein the compound is selected from the following table:
  • the invention features a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier.
  • the invention features a method of inhibiting a voltage-gated sodium ion channel in:
  • the voltage-gated sodium ion channel is NaV 1.7.
  • the invention features a method of treating or lessening the severity in a subject of acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpatic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders, anxiety, depression, dipolar disorder, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, cancer pain, stroke, cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, stressor exercise induced angina, palpitations, hypertension, migraine, or abormal gastro- intestinal mot
  • the method is used for treating or lessening the severity of femur cancer pain; non-malignant chronic bone pain; rheumatoid arthritis; osteoarthritis; spinal stenosis; neuropathic low back pain; neuropathic low back pain; myofascial pain syndrome; fibromyalgia; temporomandibular joint pain; chronic visceral pain, abdominal pain; pancreatic; IBS pain; chronic and acute headache pain; migraine; tension headache, including, cluster headaches; chronic and acute
  • neuropathic pain post-herpatic neuralgia; diabetic neuropathy; HIV-associated neuropathy; trigeminal neuralgia; Charcot-Marie Tooth neuropathy; hereditary sensory neuropathies; peripheral nerve injury; painful neuromas; ectopic proximal and distal discharges; radiculopathy; chemotherapy induced neuropathic pain; radiotherapy- induced neuropathic pain; post-mastectomy pain; central pain; spinal cord injury pain; post-stroke pain; thalamic pain; complex regional pain syndrome; phantom pain;
  • intractable pain intractable pain; acute pain, acute post-operative pain; acute musculoskeletal pain; joint pain; mechanical low back pain; neck pain; tendonitis; injury/exercise pain; acute visceral pain, abdominal pain; pyelonephritis; appendicitis; cholecystitis; intestinal obstruction; hernias; chest pain, cardiac pain; pelvic pain, renal colic pain, acute obstetric pain, labor pain; cesarean section pain; acute inflammatory, burn and trauma pain; acute intermittent pain, endometriosis; acute herpes zoster pain; sickle cell anemia; acute pancreatitis; breakthrough pain; orofacial pain including sinusitis pain, dental pain; multiple sclerosis (MS) pain; pain in depression; leprosy pain; Behcet's disease pain; adiposis dolorosa; phlebitic pain; Guillain-Barre pain; painful legs and moving toes; Haglund syndrome;
  • the compounds of the invention may be prepared readily using the following methods. Illustrated below in Scheme 1 through Scheme 21 are methods for preparing the compounds of the invention.
  • Acid, coupling reagent e.g. EDCI or HATU
  • base e.g. triethylamine or diisopropylethylamine
  • solvent e.g. DMF or CH 2 C1 2
  • base e.g. NaHC03 or NaH, diisopropylethylamine
  • solvent e.g. DMF or CH 2 C1 2
  • R 2 X (X CHO): reducing agent (e.g.
  • R 1 optionally substituted phenyl
  • PG 1 acid-stable protecting group (e.g. PMB, benzyl)
  • PG 2 acid-labile protecting group (e.g. Boc);
  • reducing agent e.g. NaBH 4 orNaCNBH 3
  • solvent e.g. MeOH
  • solvent e.g. MeOH
  • solvent e.g. EtOH
  • reflux e.g. HBr
  • H + protic acid
  • PG 1 Boc: Boc 2 0, base (e.g. Et 3 N), sqlvent (e.g. DCM)
  • PG 2 benzyl: catalyst (e.g. Pd/C or Pd(OH) 2 /C), hydrogen source (e.g. H 2 or ammonium formate), solvent (e.g. MeOH, EtOH or iPrOH);
  • base e.g.
  • Amine e.g. benzylamine
  • solvent e.g. methanol, ethanol, isopropanol
  • catalyst e.g. Pd(PPh 3 ) 4
  • ligand e.g. PPh 3
  • Z -but-2-ene-l,4-diyl diacetate
  • base e.g. triethylamine, diisopropylethylamine
  • solvent e.g. THF, diethyl ether, dioxane
  • Pd 2 (dba) 3 or Pd(OAc) 2 ligand (e.g. rac-BINAP or DPPF), base (NaOtBu or KOH), solvent (e.g. toluene or dioxane).
  • ligand e.g. rac-BINAP or DPPF
  • base NaOtBu or KOH
  • solvent e.g. toluene or dioxane
  • R 2 -X leaving group: halo, OTs, OTf
  • base e.g. K 2 C0 3 , NaHC0 3 , Et 3 N
  • solvent e.g. THF, DCM, EtOH, CH 3 CN, DMF
  • H + e.g. HCl or TFA
  • A-C0 2 H A-C0 2 H, coupling agent (e.g. HATU or EDCI), base (e.g. Et 3 N or iPr 2 NEt), solvent (e.g.
  • R 2 -X leaving group: halo, OTs, OTf
  • base e.g. K 2 C0 3 , NaHC0 3 , Et 3 N
  • solvent e.g. THF, DCM, EtOH, CH 3 CN, DMF
  • reductive amination conditions e.g. NaBH(OAc) 3 , DCE, AcOH, TEA, appropriate ketone or aldehyde
  • nucelophilic substitution conditions e.g. aryl halide, DMSO, heat
  • Pd-mediated amine arylation conditions catalyst (e.g. Pd 2 (dba) 3 or Pd(OAc) 2 ), ligand (e.g. rac-BINAP or DPPF), base
  • solvent e.g. toluene or dioxane
  • Oxidant e.g. Os0 4 , NMO
  • solvent e.g. acetone/water
  • Oxidant e.g. NaI0 4
  • solvent e.g. THF/water
  • Oxidant e.g. NaC10 2
  • solvent e.g. tBuOH/water
  • alkylating agent e.g. Mel, dimethyl sulfate
  • base e.g. 2 C0 3 , NaHC0 3 , Et 3 N
  • solvent e.g. THF, DMF
  • Grignard reagent or alkyllithium e.g. methylmagnesium halide or methyllithium
  • solvent e.g.
  • Protected amine e.g. N-benzyl-2-methylprop-2-en-l -amine
  • solvent e.g. methanol, ethanol, isopropanol
  • I 2 solvent
  • solvent e.g. H 2 0, DCM, MTBE, diethyl ether, THF
  • base e.g. NaHC0 3 , K 2 C0 3 , Et 3 N
  • Reductant e.g.
  • R -X leaving group: halo, OTs, OTf
  • base e.g. K 2 C0 3 , NaHC0 3 , Et 3 N
  • solvent e.g. THF, DCM, EtOH, CH 3 CN, DMF
  • Base e.g. NaOH, 2 C0 3 , Na 2 C0 3
  • solvent e.g. MeOH, EtOH, H 2 0.
  • Amine e.g. / methoxybenzylamine
  • solvent e.g. methanol, ethanol, isopropanol
  • 2-halo-2-arylacetyl chloride base (e.g. Et 3 N, ; ' -PrNEt 2 , pyridine), solvent (e.g. THF, DCM, CH 3 C );
  • Base e.g. NaH, Oi-Bu, NaOt-Bu
  • solvent e.g.
  • oxidant e.g. CAN or DDQ
  • solvent e.g. H 2 0/CH 3 CN or DCM, DMF, dioxane
  • PG 2 Boc
  • H + e.g. HC1 or TFA
  • A- C0 2 H coupling agent (e.g. HATU or EDCI), base (e.g. Et 3 N or iPr 2 NEt), solvent (e.g.
  • Aldehyde e.g. 3-methylbutanal
  • reductant e.g. NaBH 4 , NaBH(OAc) 3 , NaBH 3 CN
  • solvent e.g. methanol, THF, DCM, DCE
  • PG 1 trifluoroacetate; trifluoracetic anhydride or ethyl trifluoroacetate, base (e.g. pyridine or Et 3 N);
  • PG 2 benzyl; catalyst (e.g.
  • Pd/C or Pd(OH) 2 /C hydrogen source (e.g. H 2 or ammonium formate), solvent (e.g. MeOH, EtOH or iPrOH,); d) A-C0 2 H, coupling agent (e.g. HATU or EDCI), base (e.g. Et 3 N or iPr 2 NEt), solvent (e.g. DMF, CH 3 CN or CH 2 C1 2 ); e) Base (e.g. NaOH, 2 C0 3 , Na 2 C0 3 ), solvent (e.g. MeOH, EtOH, H 2 0); f) 2-halo-2- arylacetyl chloride, base (e.g.
  • PG acid-labile protecting group (e.g. Boc).
  • a) ⁇ , ⁇ -dimethylhydroxylamine, base e.g. LiHMDS, NaHMDS, LDA
  • solvent e.g. THF, diethyl ether
  • b) alkynylmagnesium halide solvent (e.g. THF, diethyl ether)
  • c) hydrazine solvent (e.g. MeOH, EtOH, z ' -PrOH)
  • R 7 -X leaving group: halo, OTs, OTf), base (e.g. NaH, KOi-Bu, NaOi-Bu), solvent (e.g.
  • a) Alkylmagnesium halide, solvent (e.g. THF, diethyl ether); b) DMF-DMA; c) hydrazine, solvent (e.g. MeOH, EtOH, /-PrOH); d) PG 1 Boc; H + (e.g. HC1 or TFA).
  • Oxidant e.g. NaI0 4
  • solvent e.g. THF/water
  • Oxidant e.g. NaC10 2
  • solvent e.g. tBuOH/water
  • base e.g. LiOH or NaOH
  • solvent e.g. THF or dioxane or MeOH
  • PG 1 acid-labile protecting group (e.g. Boc)
  • PG' acid-stable protecting group (e.g. benzyl)
  • Activating/dehydrating reagent e.g. T 3 P, HATU, EDCI
  • base e.g. Et 3 N
  • solvent e.g. 2-methyltetrahydorfuran, DMF
  • PG 1 benzyl
  • i. 1 -chloroethyl chloroformate solvent (e.g. DCE); ii. MeOH
  • R 2 -X leaving group: halo, OTs, OTf), base (e.g. K 2 C0 3 , NaHC0 3 , Et 3 N), solvent (e.g. EtOH, CH 3 CN).
  • PG acid-labile protecting group (e.g. Boc)
  • Coupling agent e.g. T3P, HATU, EDCI
  • base e.g. Et 3 N or DIEA
  • solvent e.g. 2- methyltetrahydorfuran, DMF or CH 3 CN
  • i. Oxidant e.g. Os0 4 , NMO
  • solvent e.g. acetone/ water
  • ii. Oxidant e.g. NaI0 4
  • solvent e.g. THF/water
  • X S, (e.g. Lawesson's reagent or P 2 S 5 ), solvent (e.g.
  • PG acid-stable protecting group (e.g. benzyl);
  • Coupling agent e.g. T 3 P, HATU, EDCI
  • base e.g. Et 3 N or DIEA
  • solvent e.g. 2- methyltetrahydrofuran, DMF or CH 3 CN
  • i. Oxidant e.g. Os0 4 , NMO
  • solvent e.g. acetone/ water
  • ii. Oxidant e.g. NaI0 4
  • solvent e.g. THF/water
  • X S, (e.g. Lawesson's reagent or P 2 S 5 ), solvent (e.g. THF or toluene);
  • X O, dehydrating reagent (e.g.
  • PG 1 stable protecting group (e.g. benzyl)
  • Oxidant e.g. Os0 4 , NMO
  • solvent e.g. acetone/water
  • Oxidant e.g. NaI0 4
  • solvent e.g. THF/water
  • ammonium hydroxide aldehyde/acyl equivalents (e.g. substituted/unsubstituted glyoxal, substituted 2-acetoxy ketones, alpha-bromo ketones), solvent (e.g. MeOH);
  • PG 1 benzyl; (e.g.
  • PG acid-labile protecting group (e.g. Boc).
  • a) ethyl 2-halo-2,2-disubstituted acetate, solvent e.g. DMF,THF, DCM, CH 3 CN);
  • base e.g. KOtBu
  • solvent e.g. THF
  • reductant e.g. borane-dimethylsulfide
  • solvent e.g. THF
  • R 2 -X leaving group: halo, OTs, OTf), base (e.g. K 2 C0 3 , NaHC0 3 , Et 3 N), solvent (e.g.
  • PG 2 THF, DCM, EtOH, CH 3 CN, DMF); c) diethylaminosulfur trifluoride, DCM; d) deprotection conditions for PG 2 : H + (e.g. HCl or TFA); e) A-C0 2 H, coupling agent (e.g. HATU or EDCI), base (e.g. Et 3 N or iPr 2 NEt), solvent (e.g. DMF, CH 3 CN or CH 2 C1 2 ).
  • H + e.g. HCl or TFA
  • A-C0 2 H A-C0 2 H
  • coupling agent e.g. HATU or EDCI
  • base e.g. Et 3 N or iPr 2 NEt
  • solvent e.g. DMF, CH 3 CN or CH 2 C1 2 .
  • compositions are provided.
  • the invention provides compounds that are inhibitors of voltage-gated sodium ion channels, and thus the present compounds are useful for the treatment of diseases, disorders, and conditions including, but not limited to acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, and incontinence.
  • diseases, disorders, and conditions including, but not limited to acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders,
  • compositions comprising any of the compounds as described herein, and optionally comprise a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • these compositions optionally further comprise one or more additional therapeutic agents.
  • a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a subject in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • a “pharmaceutically acceptable salt” means any non-toxic salt or salt of an ester of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • inhibitors as used herein, the term "inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of a voltage-gated sodium ion channel.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • compositions of this invention include those derived from suitable inorganic and organic acids and bases. Examples of
  • nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate,
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci- 4 alkyl)4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • the pharmaceutically acceptable compositions of the invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable carrier, adjuvant, or vehicle which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and
  • any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
  • materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymefhyl cellulose, ethyl cellulose and cellulose acetate;
  • powdered tragacanth malt; gelatin; talc; excipients such as cocoa butter and
  • suppository waxes oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
  • oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean
  • a method for the treatment or lessening the severity of acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, dipolar disorder, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, or cancer pain comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound to a subject in need thereof.
  • a method of treatment or lessening the severity of stroke, cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, stress- or exercise induced angina, palpitations, hypertension, migraine, or abormal gastro-intestinal motility comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound to a subject in need thereof.
  • a method for the treatment or lessening the severity of acute, chronic, neuropathic, or inflammatory pain is provided comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
  • a method for the treatment or lessening the severity of radicular pain, sciatica, back pain, head pain, or neck pain comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
  • a method for the treatment or lessening the severity of severe or intractable pain, acute pain, postsurgical pain, back pain, tinnitis or cancer pain is provided comprising administering an effective amount of a compound or a
  • hereditary sensory neuropathies peripheral nerve injury; painful neuromas; ectopic proximal and distal discharges; radiculopathy; chemotherapy induced neuropathic pain; radiotherapy-induced neuropathic pain; post-mastectomy pain; central pain; spinal cord injury pain; post-stroke pain; thalamic pain; complex regional pain syndrome; phantom pain; intractable pain; acute pain, acute post-operative pain; acute musculoskeletal pain; joint pain; mechanical low back pain; neck pain; tendonitis; injury/exercise pain; acute visceral pain, including, abdominal pain; pyelonephritis; appendicitis;
  • cholecystitis cholecystitis
  • intestinal obstruction hernias; etc
  • chest pain including, cardiac Pain
  • pelvic pain renal colic pain
  • acute obstetric pain including, labor pain
  • cesarean section pain acute inflammatory, burn and trauma pain
  • acute intermittent pain including, endometriosis
  • acute herpes zoster pain sickle cell anemia
  • pancreatitis breakthrough pain; orofacial pain including sinusitis pain, dental pain; multiple sclerosis (MS) pain; pain in depression; leprosy pain; behcet's disease pain; adiposis dolorosa; phlebitic pain; Guillain-Barre pain; painful legs and moving toes; Haglund syndrome; erythromelalgia pain; Fabry's disease pain; bladder and urogenital disease, including, urinary incontinence; hyperactivity bladder; painful bladder syndrome; interstitial cyctitis (IC); or prostatitis; complex regional pain syndrome (CRPS), type I and type II; angina-induced pain is provided, comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
  • MS multiple sclerosis
  • an "effective amount" of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of one or more of acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, tinnitis or cancer pain.
  • the compounds and compositions, according to the method of the invention may be administered using any amount and any route of administration effective for treating or lessening the severity of one or more of acute, chronic, neuropathic, or inflammatory pain, arthritis, migraine, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, tinnitis or cancer pain.
  • trigeminal neuralgia such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendoc
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • the compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the subject to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular subject or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • subject or "patient”, as used herein, means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3- butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary am
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in microencapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, eardrops, and eye drops are also examples of suitable pharmaceutically acceptable carrier.
  • the invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • dosage forms are prepared by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the compounds of the invention are useful as inhibitors of voltage- gated sodium ion channels.
  • the compounds and compositions of the invention are inhibitors of one or more of NaVl . l, NaV1.2, NaV1.3, NaV1.4, NaV1.5, NaV1.6, NaV1.7, NaVl .8, or NaV1.9, and thus, without wishing to be bound by any particular theory, the compounds and
  • compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder where activation or hyperactivity of one or more of NaVl .1 , NaV1.2, NaVl .3, NaV1.4, NaV1.5, NaV1.6, NaV1.7, NaV1.8, or NaV1.9 is implicated in the disease, condition, or disorder.
  • NaVl .1, NaV1.2, NaVl .3, NaVl .4, NaVl .5, NaVl .6, NaVl .7, NaVl .8, or NaVl .9 is implicated in a particular disease, condition, or disorder
  • the disease, condition, or disorder may also be referred to as a "NaVl .l, NaVl .2, NaVl .3, NaVl .4, NaVl .5, NaVl .6, NaVl .7, NaVl .8 or NaV1.9-mediated disease, condition or disorder”.
  • the invention provides a method for treating or lessening the severity of a disease, condition, or disorder where activation or hyperactivity of one or more of NaVl . l , NaVl .2, NaVl .3, NaVl .4, NaVl .5, NaVl .6, NaVl .7, NaVl .8, or NaVl .9 is implicated in the disease state.
  • the activity of a compound utilized in this invention as an inhibitor of NaVl .1 , NaVl .2, NaVl .3, NaVl .4, NaVl .5, NaVl .6, NaVl .7, NaVl .8, or NaVl .9 may be assayed according to methods described generally in the Examples herein, or according to methods available to one of ordinary skill in the art.
  • compounds of the invention are useful as inhibitors of NaV1.7 and/or NaV1.8.
  • the compounds and pharmaceutically acceptable compositions of the invention can be employed in combination therapies, that is, the compounds and pharmaceutically acceptable compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures.
  • the particular combination of therapies can be employed in combination therapies, that is, the compounds and pharmaceutically acceptable compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures.
  • therapeutics or procedures to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder (for example, an inventive compound may be administered concurrently with another agent used to treat the same disorder), or they may achieve different effects (e.g., control of any adverse effects).
  • additional therapeutic agents that are normally administered to treat or prevent a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated".
  • exemplary additional therapeutic agents include, but are not limited to: nonopioid analgesics (indoles such as Etodolac, Indomethacin,
  • Piroxicam para-aminophenol derivatives, such as Acetaminophen
  • propionic acids such as Fenoprofen, Flurbiprofen, Ibuprofen, Ketoprofen, Naproxen, Naproxen sodium, Oxaprozin
  • salicylates such as Asprin, Choline magnesium trisalicylate
  • fenamates such as meclofenamic acid, Mefenamic acid
  • pyrazoles such as Phenylbutazone
  • opioid (narcotic) agonists such as Codeine, Fentanyl
  • nondrug analgesic approaches may be utilized in conjunction with administration of one or more compounds of the invention.
  • anesthesiologic intraspinal infusion, neural blocade
  • neurosurgical aspinal infusion, neural blocade
  • neurosurgical aspinal infusion, neural blocade
  • neurostimulatory transcutaneous electrical nerve stimulation, dorsal column stimulation
  • physiatric physical therapy, orthotic devices, diathermy
  • psychologic psychologic
  • additional appropriate therapeutic agents are selected from the following:
  • an opioid analgesic e.g. morphine, heroin, hydromorphone, oxymorphone, levorphanol, levallorphan, methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, oxycodone, hydrocodone, propoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine or pentazocine;
  • opioid analgesic e.g. morphine, heroin, hydromorphone, oxymorphone, levorphanol, levallorphan, methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, oxycodone, hydrocodone, propoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nal
  • NSAID nonsteroidal antiinflammatory drug
  • diclofenac diflusinal, etodolac
  • fenbufen fenoprofen
  • flufenisal flurbiprofen
  • ibuprofen indomethacin
  • ketoprofen ketorolac
  • meclofenamic acid mefenamic acid
  • meloxicam nabumetone, naproxen, nimesulide, nitroflurbiprofen, olsalazine, oxaprozin, phenylbutazone, piroxicam, sulfasalazine, sulindac, tolmetin or zomepirac
  • NSAID nonsteroidal antiinflammatory drug
  • a barbiturate sedative e.g. amobarbital, aprobarbital, butabarbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, phenobartital, secobarbital, talbutal, theamylal or thiopental;
  • a benzodiazepine having a sedative action e.g. chlordiazepoxide, clorazepate, diazepam, flurazepam, lorazepam, oxazepam, temazepam or triazolam;
  • an Hi antagonist having a sedative action e.g. diphenhydramine, pyrilamine, promethazine, chlorpheniramine or chlorcyclizine;
  • a sedative such as glutethimide, meprobamate, methaqualone or dichloralphenazone
  • a skeletal muscle relaxant e.g. baclofen, carisoprodol
  • an NMDA receptor antagonist e.g. dextromethorphan ((+)-3- hydroxy-N- methylmorphinan) or its metabolite dextrorphan ((+)-3-hydroxy-N- methylmorphinan), ketamine, memantine, pyrroloquinoline quinine, cis-4- (phosphonomethyl)-2- piperidinecarboxylic acid, budipine, EN-3231 (MorphiDex(R), a combination formulation of morphine and dextromethorphan), topiramate, neramexane or perzinfotel including an NR2B antagonist, e.g.
  • an alpha-adrenergic e.g. doxazosin, tamsulosin, clonidine, guanfacine, dexmetatomidine, modafinil, or 4-amino-6,7-dimethoxy-2-(5-methane- sulfonamido-1, 2,3,4- tetrahydroisoquinol-2-yl)-5-(2-pyridyl) quinazoline;
  • a tricyclic antidepressant e.g. desipramine, imipramine, amitriptyline or nortriptyline;
  • an anticonvulsant e.g. carbamazepine, lamotrigine, topiratmate or valproate;
  • a tachykinin (NK) antagonist particularly an NK-3, NK-2 or NK-I antagonist, e.g. ([alpha]R,9R)-7-[3,5-bis(trifluoromethyl)benzyl]-8,9, 10,11 - tetrahydro-9-methyl-5-(4- methylphenyl)-7H-[l ,4]diazocino[2,l-g][l,7]-naphthyridine- 6-13-dione (TAK-637), 5- [[(2R,3S)-2-[(lR)-l-[3,5-bis(trifluoromethyl)phenyl]ethoxy- 3-(4-fluorophenyl)-4-morpholinyl]-methyl]-l,2-dihydro-3H-l,2,4-triazol-3-one (MK- 869), aprepitant, lanepitant, dapitant or 3-[[2-methoxy-5
  • a COX-2 selective inhibitor e.g. celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib, or lumiracoxib;
  • a coal-tar analgesic in particular paracetamol
  • a neuroleptic such as droperidol, chlorpromazine, haloperidol, perphenazine, thioridazine, mesoridazine, trifluoperazine, fluphenazine, clozapine, olanzapine, risperidone, ziprasidone, quetiapine, sertindole, aripiprazole,
  • a vanilloid receptor agonist e.g. resinferatoxin
  • antagonist e.g. capsazepine
  • a beta-adrenergic such as propranolol
  • a local anaesthetic such as mexiletine
  • a corticosteroid such as dexamethasone
  • a 5-HT receptor agonist or antagonist particularly a 5-HTi B/I D agonist such as eletriptan, sumatriptan, naratriptan, zolmitriptan or rizatriptan;
  • a 5-HT2A receptor antagonist such as R(+)-alpha-(2,3-dimethoxy- phenyl)-l-[2-(4- fluorophenylethyl)]-4-piperidinemethanol (MDL- 100907);
  • MDL- 100907 a 5-HT2A receptor antagonist
  • a cholinergic (nicotinic) analgesic such as ispronicline (TC-
  • a PDEV inhibitor such as 5-[2-ethoxy-5-(4-methyl-l-piperazinyl- sulphonyl)phenyl]- l-methyl-3-n-propyl-l,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7- one (sildenafil), (6R, 12aR)- 2,3,6,7, 12,12a-hexahydro-2-methyl-6-(3, 4- methylenedioxyphenyl)-pyrazino[2',r:6,l]- pyrido[3,4-b]indole-l,4-dione (IC-351 or tadalafil), 2-[2-ethoxy-5-(4-ethyl-piperazin-l-yl-l- sulphonyl)-phenyl]-5-methyl-7- propyl-3H-imidazo[5,l-f][l,2,4]triazin-4-one
  • a serotonin reuptake inhibitor such as sertraline, sertraline metabolite demethylsertraline, fluoxetine, norfluoxetine (fluoxetine desmethyl metabolite), fluvoxamine, paroxetine, citalopram, citalopram metabolite
  • a noradrenaline (norepinephrine) reuptake inhibitor such as maprotiline, lofepramine, mirtazepine, oxaprotiline, fezolamine, tomoxetine, mianserin, buproprion, buproprion metabolite hydroxybuproprion, nomifensine and viloxazine (Vivalan(R)), especially a selective noradrenaline reuptake inhibitor such as reboxetine, in particular (S,S)-reboxetine;
  • a dual serotonin-noradrenaline reuptake inhibitor such as venlafaxine, venlafaxine metabolite O-desmethylvenlafaxine, clomipramine, clomipramine metabolite desmethylclomipramine, duloxetine, milnacipran and imipraniine;
  • a dual serotonin-noradrenaline reuptake inhibitor such as venlafaxine, venlafaxine metabolite O-desmethylvenlafaxine, clomipramine, clomipramine metabolite desmethylclomipramine, duloxetine, milnacipran and imipraniine
  • iNOS inducible nitric oxide synthase
  • a prostaglandin E2 subtype 4 (EP4) antagonist such as N-[( ⁇ 2-[4- (2-ethyl-4,6- dimethyl-lH-imidazo[4,5-c]pyridin-l-yl)phenyl]ethyl ⁇ amino)-carbonyl]-4- methylbenzenesulfonamide or 4-[(l 5)-l-( ⁇ [5-chloro-2-(3-fluorophenoxy)pyridin-3- yljcarbonyl ⁇ amino)ethyl]benzoic acid;
  • a leukotriene B4 antagonist such as l-(3-biphenyl-4-ylmethyl-4- hydroxy-chroman-7- yl)-cyclopentanecarboxylic acid (CP- 105696), 5-[2-(2- Carboxyethyl)-3-[6-(4- methoxyphenyl)-5E- hexenyljoxyphenoxy] -valeric acid ( ⁇ - 4057) or DPC-11870, [00170] (35) a 5-lipoxygenase inhibitor, such as zileuton, 6-[(3-fluoro-5-[4- methoxy-3,4,5,6- tetrahydro-2H-pyran-4-yl])phenoxy-methyl]-l-methyl-2-quinolone (ZD-2138), or 2,3,5- trimethyl-6-(3-pyridylmethyl),l ,4-benzoquinone (CV-6504);
  • a sodium channel blocker such as lidocaine
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the invention in another aspect, includes a composition for coating an implantable device comprising a compound of the invention as described generally above, and in classes and subclasses herein, and a carrier suitable for coating said implantable device.
  • the invention includes an implantable device coated with a composition comprising a compound of the invention as described generally above, and in classes and subclasses herein, and a earner suitable for coating said implantable device.
  • Suitable coatings and the general preparation of coated implantable devices are described in US Patents 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Another aspect of the invention relates to inhibiting one or more of NaVl . l , NaV1.2, NaV1.3, NaVl .4, NaVl .5, NaVl .6, NaVl .7, NaVl .8, or NaVl .9, activity in a biological sample or a subject, which method comprises administering to the subject, or contacting said biological sample with a compound of formula I or a composition comprising said compound.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of one or more of NaVl .1 , NaVl .2, NaVl .3, NaVl .4, NaVl .5, NaVl .6, NaVl .7, NaVl .8, or NaVl .9, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, the study of sodium ion channels in biological and pathological phenomena; and the comparative evaluation of new sodium ion channel inhibitors.
  • the LC/MS eluting system was 1-99% or 10-99% acetonitrile in H 2 0 with 0.035% v/v trifluoroacetic acid, 0.035% v/v formic acid, 5 mM HC1 or 5 mM ammonium formate using a 3 or 15 minute linear gradient and a flow rate of 12 mL/minute.
  • Silica gel chromatography was performed using silica gel-60 with a particle size of 230-400 mesh. Pyridine, dichloromethane (CH 2 C1 2 ), tetrahydrofuran (THF),
  • DMF dimethylformamide
  • ACN acetonitrile
  • MeOH methanol
  • 1,4-dioxane was from Aldrich Sure-Seal bottles kept under dry nitrogen. All reactions were stirred magnetically unless otherwise noted.
  • Step 2 [00182] A solution of 2-(benzylamino)-l -phenyl ethanol (5.3 g, 23.44 mmol) and tert-butyl l-oxa-6-azaspiro[2.5]octane-6-carboxylate (5.0 g, 23.44 mmol) in ethanol (30 mL) was heated overnight at 75 °C. The solvent was evaporated and the crude material was used in the next step without further purification. ESI-MS m/z calc. 440.2, found 441.7 (M+l) + ; Retention time: 1.41 minutes (3 min run).
  • Step 2 [00218] To a solution of tert- uXyl 4-[[benzyl(2-methylallyl)amino]methyl]-4- hydroxy-piperidine-l-carboxylate (1.2 g, 3.20 mmol) in 2-methoxy-2-methyl -propane (1 1 mL) was added aq. NaHC0 3 (4.3 mL of 1 M, 4.30 mmol) then iodine (0.9 g, 181 ⁇ , 3.52 mmol) and the reaction mixture was rapidly stirred for 16 hours.
  • reaction mixture was diluted with ethyl acetate (25 mL), quenched with 5 mL 1 M Na 2 S 2 0 3 aq., shaken vigorously and separated. The organic layer was washed with 1 : 1 sat. aq.
  • reaction mixture was heated at 60 °C for 5 hours, then at room temperature for 72 hours, then microfiltered and purified by prep LCMS (1-99% ACN/Water, 5 mM HCl modifier) to give l-(8-ethyl-10,10-dim ethyl- 1 l-oxa-3,8-diazaspiro[5.5]undecan-3-yl)-2,2,2- trifluoro-ethanone hydrochloride salt (47 mg, 52 %) as a white solid.
  • ESI-MS m/z calc. 308.2, found 309.7 (M+l) + ; Retention time: 0.90 minutes (3 min run).
  • Step 2 [00234] To teri-butyl 10-benzyl-8-vinyl-7-oxa-3,10-diazaspiro[5.5]undecane-3- carboxylate (2.07 g, 5.56 mmol) and 4-methylmorpholine 4-oxide (725 mg, 6.19 mmol) in acetone (19 mL) and water (2 mL) was added osmium tetroxide in water (704 ⁇ L of 2.5 %w/w, 0.06 mmol) dropwise and the solution was stirred for 2 hours.
  • the reaction mixture was quenched with 1M sodium thiosulfate (50 mL) and stirred for 5 minutes, then extracted with EtOAc (4 x 50 mL), washed with sat. aq. NaHC0 3 (30 mL), and dried over MgS0 4 , filtered and concentrated in vacuo to give the diol intermediate.
  • the diol was diluted with DCM (20 mL), filtered over a plug of neutral alumina, washing extensively with 20% MeOH/DCM (> 1 L).
  • reaction mixture was concentrated in vacuo then purified by silica gel column chromatography using 0-70% EtOAc/hexane as eluent to give 9-tert- butyl 2-methyl 4-benzyl-l-oxa-4,9-diazaspiro[5.5]undecane-2,9-dicarboxylate (1 g, 67%) as a colorless oil.
  • Step 6 [00242] 9-73 ⁇ 4 -butyl 2-methyl 4-(2,2,2-trifluoroethyl)-l -oxa-4,9- diazaspiro[5.5]undecane-2,9-dicarboxylate (197 mg, 0.48 mmol) was dissolved in MeOH (1 mL) / H 2 0 (1 mL), followed by the addition of LiOH (46 mg, 1.92 mmol) and the reaction mixture was stirred for 2 hours at room temperature.
  • Step 2 [00253] To tert-butyl 4-(2,2-difluoroethyl)-2-((2-oxobutyl)carbamoyl)-l -oxa-
  • Step 5 [00272] HCl (781 ⁇ , of 4 M in dioxane, 3.12 mmol) was added to a solution of tert-butyl 4-(2,2-difluoroethyl)-2-(oxazol-2-yl)-l-oxa-4,9-diazaspiro[5.5]undecane-9- carboxylate (121 mg, 0.31 mmol) in DCM (0.7 mL) and the reaction mixture was stirred for 1 hour at room temperature.
  • T3P (591 mg, 553 ⁇ of 50 %w/w, 0.93 mmol) was added to a mixture of 9-(teri-butoxycarbonyl)-4-(2,2,2-trifluoroethyl)- 1 -oxa-4,9-diazaspiro[5.5]undecane- 2-carboxylic acid (142 mg, 0.37 mmol), l-aminopropan-2-one hydrochloride salt (41 mg, 0.37 mmol) and Et 3 N (188 mg, 259 ⁇ ,, 1.86 mmol) in 2-methyltetrahydrofuran (706 ⁇ The reaction mixture was then heated at 45°C for 2 hours.
  • the reaction mixture was quenched with sat. aq. NH 4 C1 (30 mL), extracted with ether (3 x 50 mL), and the combined organics were dried over MgS0 4 , filtered and concentrated in vacuo.
  • the crude material was purified by silica gel column chromatography (0-100% ethyl acetate/dichloromethane) to give tert-buty ⁇ 8-(2,2-difiuoroethyl)-10- [methoxy(methyl)carbamoyl]-l l-oxa-3,8-diazaspiro[5.5]undecane-3-carboxylate (1.38 g, 85%) as a colorless oil.
  • Step 2 [00286] To tert-butyl 8-(2,2-difluoroethyl)-10-[methoxy(methyl)carbamoyl]-
  • Step 2 [00295] To tert-butyl 4-[[benzyl-[2-oxo-2-(2-pyridyl)ethyl]amino]methyl]-4- hydroxy-piperidine-1 -carboxylate (190 mg, 0.43 mmol) in benzene (11 mL) was added 4-methylbenzenesulfonic acid (99 mg, 0.52 mmol) and reaction mixture was heated at 80 °C for 30 minutes. The reaction mixture was cooled, diluted with ethyl acetate and washed sequentially with sat. NaHC0 3 and brine solution.
  • Step 3 [00297] To ieri-butyl 8-benzyl-10-(2-pyridyl)-l l-oxa-3,8- diazaspiro[5.5]undec-9-ene-3-carboxylate (80 mg, 0.19 mmol) in methanol (13 mL) was added Pd(OH) 2 (50 mg, 0.36 mmol) and ammonium formate (180 mg, 2.85 mmol) and the reaction mixture was heated at 70 °C for 1 hour. The reaction mixture was cooled, diluted with ethyl acetate and filtered through Celite®. The organic layer was washed with 1 : 1 NaOH:NaHC0 3 solution, dried over Na 2 S0 4 , filtered and
  • Step 1 A solution of tert-butyl-l-oxa-6-azaspiro[2.5]octane-6-carboxylate (0.7 g, 3.42 mmol) and 2-[(4-methoxyphenyl)methylamino]-2-phenyl-ethanol (800 mg, 3.109 mmol) in ethanol (5 mL) was stirred at 60 °C for 72 hours.
  • reaction mixture was concentrated in vacuo and purified by silica gel column chromatography using 0 to 10% MeOH in DCM as eluent to yield tert-butyl 4-hydroxy-4-[[(2-hydroxy-l -phenyl- ethyl)-[(4-methoxyphenyl)methyl]amino]methyl]piperidine-l -carboxylate (1.2 g, 82 %).
  • ESI-MS m/z calc. 470.6, found 471.5 (M+l) + ; Retention time: 1.45 minutes (3 min run).
  • Step 2 [00317] To a solution of teri-butyl 4-hydroxy-4-[[(2-hydroxy-l -phenyl-ethyl)-
  • Step 5 [00336] Tert-butyl 10-(methoxymethyl)-8-pyrimidin-2-yl-l l-oxa-3,8- diazaspiro[5.5]undecane-3-carboxylate (1.18 g, 3.12 mmol) was dissolved in dichloromethane (2 mL) and treated with a solution of HC1 (1.6 mL of 4 M in dioxane, 6.2 mmol). The mixture was allowed to stir for 2 h. The reaction mixture was evaporated to dryness to provide 2-(methoxymethyl)-4-(pyrimidin-2-yl)-l-oxa-4,9- diazaspiro[5.5] undecane hydrochloride.
  • Step 1 To a solution of teri-butyl 8-benzyl-10-(hydroxymethyl)-l l-oxa-3,8- diazaspiro[5.5]undecane-3-carboxylate (2.00 g, 5.31 mmol) in methanol (38 mL) was added Pd(OH) 2 (671 mg, 4.78 mmol) and ammonium formate (4.02 g, 63.7 mmol) and the mixture heated at 50 °C for 1 h. Additional catalyst (2.6 mmol) and ammmonium formate (25 mmol) were added and the reaction heated for an additional 2 h. The reaction mixture was filtered and concentrated to ⁇ 10 mL volume.
  • the concentrate was diluted with dichloromethane and saturated sodium bicarbonate solution.
  • the organic layer was separated and the aqueous layer washed with dichloromethane (5 ⁇ 50 mL).
  • the combined organics were dried over Na 2 S0 4 , filtered, and concentrated to provide tert-butyl 10-(hydroxymethyl)-l l-oxa-3,8-diazaspiro[5.5]undecane-3- carboxylate (1.37 g, 4.77 mmol, 90%) a s an amber-colored glass.
  • ESI-MS m/z calc. 286.2, found 287.3 (M+l) + ; Retention time: 0.65 minutes (3 min run).
  • the reaction mixture was filtered through Celite and rinsed with tetrahydrofuran (30 mL). The filtrate solution was cooled to 0 °C and treated portion- wise with sodium borohydride (223 mg, 5.91 mmol) over 5 min. The reaction mixture was stirred for 30 min then diluted with brine (400 mL) and ethyl acetate (400 mL). The organic phase was separated and the aqueous phase extracted with ethyl acetate (3 x 100 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo.
  • Step 3 To a solution of tert-butyl 8-terf-butyl- 10-(hydroxymethyl)- 1 1 -oxa-3 ,8- diazaspiro[5.5]undecane-3-carboxylate (720 mg, 2.10 mmol) in N,N- dimethylformamide (7.0 mL) was added sodium hydride (588 mg, 14.7 mmol) and the reaction stirred for 10 min. Methyl iodide (916 i , 14.7 mmol) was added and the reaction mixture stirred for 20 min. The reaction was quenched with saturated aqueous NH 4 C1 and extracted with ethyl acetate (2 x 25 mL).
  • Step 4 [00375] Tert-butyl 8-tert-butyl-10-(methoxymethyl)-l l-oxa-3,8- diazaspiro[5.5]undecane-3-carboxylate (732 mg, 2.05 mmol) was dissolved in dichloromethane (10 mL) and treated dropwise with a solution of HCl in dioxane (12.8 mL of 4 M, 51.3 mmol). The reaction was stirred for 1.5 h and then diluted with dichloromethane.
  • Step 1 Step 1 :
  • the organic layer was discarded and then the aqueous layer was acidified with 4M aqueous hydrochloric acid.
  • the aqueous layer was then extracted 3 times with ethyl acetate.
  • the combined extracts were dried over sodium sulfate and then concentrated in vacuo.
  • the crude material was purified by silica gel column chromatography using a gradient of 0-10% methanol in dichloromethane to give 4-(l -hydroxy- l-methyl-ethyl)-3 -methyl - benzoic acid (1.51 g, 42%).
  • dichloromethane (5 mL). The reaction mixture was allowed to stir at room temperature for 24 hours. Water (5 mL) and dichloromethane (5 mL) were added and the two phases were separated. The aqueous layer was extracted with dichloromethane (3 x 5 mL) and the combined organics were stirred with 10% aqueous potassium hydroxide until all remaining starting material was not observed in the organic phase (TLC, 40% ethyl acetate in hexanes).
  • triphenylphosphine (3.17 g, 2.8 mL, 12.1 mmol) in THF (63.28 mL) at 0 °C was added DIAD (2.44 g, 2.34 mL, 12.1 mmol). The ice bath was removed and the reaction was stirred at 55 °C for 16 hours. The reaction mixture was diluted with EtOAc and washed sequentially with NaHC0 3 (2 x 20 mL) and brine (2 x 20 mL) solutions. The organic layer was separated, dried over Na 2 S0 4 , filtered and the solvent was evaporated under reduced pressure.
  • Step 2 [00408] To methyl 3-methoxy-4-(2-methylallyloxy)benzoate (313 mg, 1.33 mmol) in MeOH (2.5 mL) was added H 2 S0 4 (71 ⁇ ⁇ , 1.3 mmol) and the reaction mixture was heated in a microwave vial at 100 °C for 15.5 hours. The reaction mixture was concentrated in vacuo and the crude product was purified by silica gel column chromatography using 0-30% ethyl acetate in hexane as eluent to yield methyl 3- methoxy-4-(2-methoxy-2-methyl-propoxy)benzoate (208 mg, 59 %).
  • ESI-MS m/z calc.
  • Butyllithium (16 mL of 1.6 M, 25.6 mmol) was added dropwise to a mixture of 4-bromo-3-methyl-benzoic acid (2.5 g, 1 1.6 mmol) and THF (63 mL) at -78 °C. The reaction mixture was allowed to stir at -78 °C for 30 minutes before a solution of 2-isopropyldisulfanylpropane (1.7 g, 11.6 mmol) in THF (2 mL) was added dropwise. The mixture was allowed to stir at -78 °C for 30 minutes, then 30 minutes at room temperature. The reaction mixture was then diluted with 1 M aqueous sodium hydroxide (100 mL).
  • Step 2 [00422] To methyl 3-chloro-4-hydroxy-benzoate (3.0 g, 16.1 mmol) in DMF (19 mL) was added potassium carbonate (8.9 g, 64.3 mmol) followed by 2-iodopropane (5.5 g, 3.2 mL, 32.2 mmol). The reaction mixture was heated at 60 °C for 1.5 hours. The reaction mixture was cooled, filtered and diluted with EtOAc and the solvent was concentrated in vacuo. The material was dissolved in EtOAc and washed with water (3 x 10 mL) and brine (1 x 10 mL).
  • Step 1 Step 1 :
  • Step 2
  • the filtrate was diluted with water (9 ml), and washed with ether (2 x 5 mL).
  • the aqueous layer was separated and cooled to 0 °C and the pH was adjusted to pH 2 by addition of 6 M HC1 solution.
  • the aqueous layer was extracted with EtOAc (3 x 30 mL).
  • the organics were separated and washed sequentially with water and brine.
  • the organic layer was separated, dried and concentrated in vacuo to give 2-fluoro-4-isopropoxy-benzoic acid (2.45 g, 67%) as a white solid.
  • Trimethylsilyl diazomethane (1 1.6 mL of 2 M in toluene, 23.2 mmol) was added dropwise to a solution of 2-(4-bromo-2-fluoro-phenyl)acetic acid (4.5 g, 19.3 mmol) in a mixture of toluene (7.65 mL)/ methanol (7.65 mL) under a nitrogen atmosphere at room temperature. The reaction mixture was then quenched with a few drops of acetic acid and the solvents were concentrated in vacuo.
  • Step 3
  • Step 4
  • Step 2 Step 2 :
  • Step 1 Step 1 :
  • dichlorobis(triphenylphosphine) complex (753 mg, 1.07 mmol) under an atmosphere of argon was added DMF (21 mL) and the reaction mixture was cooled to 0 °C.
  • Triethylamine (1.95 mL, 13.9 mmol) was added followed by 3-methoxyprop-l-yne (997 ⁇ , 11.8 mmol) and the reaction mixture was stirred at 60 °C for 70 minutes.
  • the reaction mixture was cooled, diluted with EtOAc and filtered over Celite®. The filtrate was washed sequentially with 1 M HC1, 10% NH 4 OH and brine.
  • Step 2
  • Step 1 Step 1 :
  • the crude ester was dissolved in DMF (20 mL) and powdered sodium thiomethoxide (2.3 g, 32.4 mmol) was added and the reaction mixture was stirred at room temperature for 1 hour and at 80 °C for 1 hour.
  • the reaction mixture was concentrated in vacuo, the residue was partitioned between 1M HC1 solution and ethyl acetate. The layers were separated and the organic was washed with 1M hydrochloric acid solution. The ethyl acetate layer was then dried over magnesium sulfate, filtered, and concentrated in vacuo to give methyl 3-methyl-4- (methylthio)benzoate.
  • ESI-MS m/z calc.
  • Step 2 [00527] To a mixture of methyl 3-methyl-4-methylsulfonyl-benzoate (5.8 g,
  • Step 1 Step 1 :
  • Step 2 [00541] To ethyl 4-(cyclopropanecarbonyl)benzoate (400 mg, 1.83 mmol) in ethanol (10 mL) was added at room temperature NaBH 4 (69 mg, 1.83 mmol) and the reaction mixture was stirred for 1 hour. The reaction mixture was then concentrated in vacuo and NaOH (1.5 g, 9.38 mmol) and dioxane (1.5 mL) were added and the reaction mixture was heated at 80 °C for 3 hours. The reaction mixture was concentrated in vacuo to half the volume and pH was adjusted to 2 with 3 N HC1.
  • Step 1 To methyl 3-fluoro-4-hydroxy-benzoate (2.0 g, 11.8 mmol) in DMF
  • Step 2
  • Step 1 Step 1 :
PCT/US2012/028882 2011-03-14 2012-03-13 Morpholine-spirocyclic piperidine amides as modulators of ion channels WO2012125613A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
MX2013010476A MX347982B (es) 2011-03-14 2012-03-13 Morfolina-piperidina espirociclica-amidas como moduladores de canales ionicos.
NZ615014A NZ615014B2 (en) 2011-03-14 2012-03-13 Morpholine-spirocyclic piperidine amides as modulators of ion channels
JP2013558104A JP6088443B2 (ja) 2011-03-14 2012-03-13 イオンチャネルのモジュレーターとしてのモルホリン−スピロ環式ピペリジンアミド
CN201280018832.5A CN103517910B (zh) 2011-03-14 2012-03-13 作为离子通道调节剂的吗啉-螺环哌啶酰胺
ES12710841.3T ES2618929T3 (es) 2011-03-14 2012-03-13 Morfolina-Spiro piperidina amida cíclica como moduladores del canal iónico
CA2829803A CA2829803A1 (en) 2011-03-14 2012-03-13 Morpholine-spirocyclic piperidine amides as modulators of ion channels
AU2012229187A AU2012229187B2 (en) 2011-03-14 2012-03-13 Morpholine-spirocyclic piperidine amides as modulators of ion channels
EP12710841.3A EP2686325B1 (en) 2011-03-14 2012-03-13 Morpholine-spirocyclic piperidine amides as modulators of ion channels

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161452538P 2011-03-14 2011-03-14
US61/452,538 2011-03-14
US201161567809P 2011-12-07 2011-12-07
US61/567,809 2011-12-07

Publications (1)

Publication Number Publication Date
WO2012125613A1 true WO2012125613A1 (en) 2012-09-20

Family

ID=45888492

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/028882 WO2012125613A1 (en) 2011-03-14 2012-03-13 Morpholine-spirocyclic piperidine amides as modulators of ion channels

Country Status (9)

Country Link
US (2) US8828996B2 (ja)
EP (1) EP2686325B1 (ja)
JP (1) JP6088443B2 (ja)
CN (1) CN103517910B (ja)
AU (1) AU2012229187B2 (ja)
CA (1) CA2829803A1 (ja)
ES (1) ES2618929T3 (ja)
MX (1) MX347982B (ja)
WO (1) WO2012125613A1 (ja)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013109521A1 (en) * 2012-01-16 2013-07-25 Vertex Pharmaceuticals Incorporated Pyran-spirocyclic piperidine amides as modulators of ion channels
US8598164B2 (en) 2010-05-06 2013-12-03 Vertex Pharmaceuticals Incorporated Heterocyclic chromene-spirocyclic piperidine amides as modulators of ion channels
WO2014120808A1 (en) 2013-01-31 2014-08-07 Vertex Parmaceuticals Incorporated Pyridone amides as modulators of sodium channels
WO2014120815A1 (en) 2013-01-31 2014-08-07 Vertex Pharmaceuticals Incorporated Quinoline and quinazoline amides as modulators of sodium channels
US8916565B2 (en) 2011-02-02 2014-12-23 Vertex Pharmaceuticals Incorporated Pyrrolopyrazine-spirocyclic piperidine amides as modulators of ion channels
WO2015089361A1 (en) 2013-12-13 2015-06-18 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
WO2015185207A1 (en) * 2014-06-02 2015-12-10 Laboratorios Del Dr. Esteve, S.A. Alkyl and aryl derivatives of 1-oxa-4,9-diazaspiro undecane compounds having multimodal activity against pain
WO2017067663A1 (en) * 2015-10-23 2017-04-27 Laboratorios Del Dr. Esteve, S.A. Oxa-azaspiro compounds having activity against pain
WO2017067665A1 (en) * 2015-10-23 2017-04-27 Laboratorios Del Dr. Esteve, S.A. Substituted morpholine derivatives having activity against pain
WO2017067664A1 (en) * 2015-10-23 2017-04-27 Laboratorios Del Dr. Esteve, S.A. Oxa-diazaspiro compounds having activity against pain
US10065971B2 (en) 2014-06-02 2018-09-04 Esteve Pharmaceuticals S.A. Amide derivatives of 1-oxa-4,9-diazaspiro undecane compounds having multimodal activity against pain
WO2018213426A1 (en) 2017-05-16 2018-11-22 Vertex Pharmaceuticals Incorporated Deuterated pyridone amides and prodrugs thereof as modulators of sodium channels
WO2019014352A1 (en) 2017-07-11 2019-01-17 Vertex Pharmaceuticals Incorporated CARBOXAMIDES AS INHIBITORS OF SODIUM CHANNELS
US10385070B2 (en) 2011-02-18 2019-08-20 Vertex Pharmaceuticals Incorporated Chroman-spirocyclic piperidine amides as modulators of ion channels
WO2020146682A1 (en) 2019-01-10 2020-07-16 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
WO2020146612A1 (en) 2019-01-10 2020-07-16 Vertex Pharmaceuticals Incorporated Esters and carbamates as modulators of sodium channels
WO2020176763A1 (en) 2019-02-27 2020-09-03 Vertex Pharmaceuticals Incorporated Dosage form comprising prodrug of na 1.8 sodium channel inhibitor
WO2020219867A1 (en) 2019-04-25 2020-10-29 Vertex Pharmaceuticals Incorporated Pyridone amide co-crystal compositions for the treatment of pain
WO2021113627A1 (en) 2019-12-06 2021-06-10 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
US11203571B2 (en) 2013-07-19 2021-12-21 Vertex Pharmaceuticals Incorporated Sulfonamides as modulators of sodium channels
WO2022256708A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Solid dosage forms and dosing regimens comprising (2r,3s,4s,5r)-4-[[3-(3,4-difluoro-2-methoxy-phenyl)-4,5-dimethyl-5-(trifluoromethyl) tetrahydrofuran-2-carbonyl]amino]pyridine-2-carboxamide
WO2022256842A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Hydroxy and (halo)alkoxy substituted tetrahydrofurans as modulators of sodium channels
WO2022256679A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamide analogs as modulators of sodium channels
WO2022256702A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran-2-carboxamides as modulators of sodium channels
WO2022256676A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran analogs as modulators of sodium channels
WO2022256622A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
US11529337B2 (en) 2018-02-12 2022-12-20 Vertex Pharmaceuticals Incorporated Method of treating pain
WO2023205778A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205468A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205465A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205463A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
EP4081509A4 (en) * 2019-12-24 2024-01-03 Zydus Lifesciences Ltd NOVEL COMPOUNDS SUITABLE FOR TREATING DYSLIPIDEMIA

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX363708B (es) 2013-10-14 2019-03-29 Eisai R&D Man Co Ltd Compuestos de quinolina selectivamente sustituidos.
AU2014334554B2 (en) 2013-10-14 2018-12-06 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
TWI685497B (zh) * 2014-06-02 2020-02-21 西班牙商伊史帝夫製藥公司 具有多重模式抗疼痛活性的1-氧雜-4,9-二氮雜螺十一烷化合物之烷基衍生物
UA121775C2 (uk) 2015-07-31 2020-07-27 Пфайзер Інк. 1,1,1-трифтор-3-гідроксипропан-2-ілкарбаматні похідні та 1,1,1-трифтор-4-гідроксибутан-2-ілкарбаматні похідні як інгібітори magl
WO2017084752A1 (en) 2015-11-16 2017-05-26 Laboratorios Del Dr. Esteve, S.A. Oxadiazaspiro compounds for the treatment of drug abuse and addiction
US10570086B2 (en) * 2016-03-17 2020-02-25 Fmc Corporation Process for converting S-enantiomer to its racemic form
CA3050625C (en) 2017-01-20 2021-07-20 Pfizer Inc. 1,1,1-trifluoro-3-hydroxypropan-2-yl carbamate derivatives as magl inhibitors
EP3571202B1 (en) 2017-01-23 2021-06-30 Pfizer Inc. Heterocyclic spiro compounds as magl inhibitors
CN111263764A (zh) 2017-10-17 2020-06-09 埃斯蒂文制药股份有限公司 (r)-9-(2,5-二氟苯乙基)-4-乙基-2-甲基-1-氧杂-4,9-二氮杂螺[5.5]十一烷-3-酮的盐
CN110452183B (zh) * 2019-09-04 2023-03-14 中国科学院上海有机化学研究所 一种手性杂环化合物的制备方法
CN113278021B (zh) * 2021-05-29 2022-09-23 天津全和诚科技有限责任公司 1,7-二氮杂螺[3.5]壬烷-7-甲酸叔丁酯及其草酸盐的制备方法

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4353901A (en) * 1981-10-19 1982-10-12 Syntex (U.S.A.) Inc. 9-(1,4-Benzodioxan-2-ylalkyl and hydroxyalkyl)-1-oxa-4,9-diazaspiro[5.5]undecan-3-ones
US5304121A (en) 1990-12-28 1994-04-19 Boston Scientific Corporation Drug delivery system making use of a hydrogel polymer coating
US5886026A (en) 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
US6099562A (en) 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US20020013325A1 (en) * 1995-09-29 2002-01-31 Fisher Matthew J. Spiro compounds as inhibitors of fibrinogen-dependent platelet aggregation
US20060052597A1 (en) * 2002-10-22 2006-03-09 Best Desmond J Aryloxyalkylamine derivatives as h3 receptor ligands
WO2010151595A1 (en) * 2009-06-26 2010-12-29 Schering Corporation Pyrrolo-benzo-1,4-diazines useful as sodium channel blockers
WO2011092198A1 (en) * 2010-01-26 2011-08-04 Boehringer Ingelheim International Gmbh 5-alkynyl-pyrimidines

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3939159A (en) 1974-05-29 1976-02-17 American Hoechst Corporation Spiro(pyrrolo (1,2-A)quinoxalines)
PT67194B (en) 1976-11-02 1979-03-23 Lilly Co Eli Process for preparing 4a-aryl-octahydro-1h-2-pyrindines
PT68934A (en) 1977-12-27 1979-01-01 Lilly Co Eli Process for preparing 4a-aryl-octahydro-1h-2-pyrindines
GB8827479D0 (en) 1988-11-24 1988-12-29 Zambeletti Spa L Novel compounds
US5206240A (en) 1989-12-08 1993-04-27 Merck & Co., Inc. Nitrogen-containing spirocycles
IL96507A0 (en) 1989-12-08 1991-08-16 Merck & Co Inc Nitrogen-containing spirocycles and pharmaceutical compositions containing them
JPH04297458A (ja) 1991-03-27 1992-10-21 Toray Ind Inc イソキノリン誘導体およびその製造方法
GB9104656D0 (en) 1991-03-05 1991-04-17 Zambeletti Spa L Pharmaceuticals
DE4341403A1 (de) 1993-12-04 1995-06-08 Basf Ag N-substituierte 3-Azabicycloalkan-Derivate, ihre Herstellung und Verwendung
CA2189634A1 (en) 1994-05-06 1995-11-16 John J. Baldwin Combinatorial dihydrobenzopyran library
US6017768A (en) 1994-05-06 2000-01-25 Pharmacopeia, Inc. Combinatorial dihydrobenzopyran library
IT1276462B1 (it) 1995-07-03 1997-10-31 Rotta Research Lab Diamidi aromatiche acide ad attivita' antigastrinica, procedimento per la loro preparazione e loro uso farmaceutico
JP4297458B2 (ja) 1999-04-16 2009-07-15 クラリオン株式会社 ナビゲーションシステム及び方法並びにナビゲーション用ソフトウェアを記録した記録媒体
US20040266802A1 (en) 1999-06-07 2004-12-30 Alain Calvet Tricyclic analgesics
US20020151712A1 (en) 1999-09-14 2002-10-17 Nan-Horng Lin 3-pyrrolidinyloxy-3'-pyridyl ether compounds useful for controlling chemical synaptic transmission
AU2001289831A1 (en) 2000-09-06 2002-03-22 Bayer Aktiengesellschaft Medicaments against viral infections
US20040014744A1 (en) 2002-04-05 2004-01-22 Fortuna Haviv Substituted pyridines having antiangiogenic activity
WO2003095427A1 (fr) 2002-05-10 2003-11-20 Taisho Pharmaceutical Co.,Ltd. Compose a noyau spiranique
BR0311749A (pt) 2002-06-11 2005-03-08 Sankyo Co Processo para produzir um composto, e, composto
WO2004037828A1 (ja) 2002-10-24 2004-05-06 Sankyo Company, Limited 光学活性なスルホキシドの製造法
WO2004054974A2 (en) 2002-12-13 2004-07-01 Smithkline Beecham Corporation Piperidine derivatives as ccr5 antagonists
CA2512886A1 (en) 2003-02-28 2004-09-10 Galderma Research & Development, S.N.C. Ligands that modulate lxr-type receptors
JPWO2004092179A1 (ja) 2003-04-14 2006-07-06 日本曹達株式会社 スピロ誘導体、製造法および抗酸化薬
FR2856685B1 (fr) 2003-06-25 2005-09-23 Merck Sante Sas Derives de thiazolylpiperidine, leurs procedes de preparation et les compositions pharmaceutiques qui les contiennent
KR20140015122A (ko) 2003-10-01 2014-02-06 아돌로 코포레이션 스피로시클릭 헤테로시클릭 유도체 및 이의 사용 방법
US20070078120A1 (en) 2003-10-21 2007-04-05 Hitoshi Ban Novel piperidine derivative
TWI350168B (en) 2004-05-07 2011-10-11 Incyte Corp Amido compounds and their use as pharmaceuticals
WO2005121090A1 (en) 2004-06-02 2005-12-22 Abbott Laboratories Substituted piperidines that have antiangiogenic activity
US7598261B2 (en) 2005-03-31 2009-10-06 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
WO2006117669A1 (en) 2005-05-03 2006-11-09 Pfizer Inc. Amide resorcinol compounds
WO2007050124A1 (en) 2005-05-19 2007-05-03 Xenon Pharmaceuticals Inc. Fused piperidine derivatives and their uses as therapeutic agents
RU2422446C2 (ru) 2005-07-19 2011-06-27 Мерк Шарп Энд Домэ Корп. Производные спирохроманона в качестве ингибиторов ацетил коэнзим а карбоксилазы (асс)
CA2621255A1 (en) 2005-09-21 2007-04-05 Incyte Corporation Amido compounds and their use as pharmaceuticals
ES2407115T3 (es) 2005-11-18 2013-06-11 Ono Pharmaceutical Co., Ltd. Compuesto que contiene un grupo básico y uso del mismo
AR057579A1 (es) 2005-11-23 2007-12-05 Merck & Co Inc Compuestos espirociclicos como inhibidores de histona de acetilasa (hdac)
US20080027072A1 (en) 2006-04-20 2008-01-31 Ampla Pharmaceuticals, Inc. Potentiation of MC4 receptor activity
CA2648831A1 (en) 2006-05-05 2007-11-15 Ulrich Reiser Spiro- (tho) benzopyran-2, 4' -piperidine-and cyclohexane derivatives as inhibitors of specific cell cycle enzymes
EP2042503B1 (en) 2006-05-16 2013-01-30 Ono Pharmaceutical Co., Ltd. Compound having acidic group which may be protected, and use thereof
CA2651681A1 (en) 2006-05-18 2007-11-29 Merck & Co., Inc. Aryl-fused spirocyclic compounds
EP2055705A4 (en) 2006-07-31 2014-08-20 Ono Pharmaceutical Co COMPOUND WITH A CYCLIC GROUP BOUND BY A SPIRO BINDING THEREOF AND APPLY THEREOF
TW200831497A (en) 2006-10-12 2008-08-01 Epix Delaware Inc Carboxamide compounds and their use
US20090270435A1 (en) 2006-11-29 2009-10-29 Jeffrey Wayne Corbett Spiroketone Acetyl-CoA Carboxylase Inhibitors
PE20081559A1 (es) 2007-01-12 2008-11-20 Merck & Co Inc DERIVADOS DE ESPIROCROMANONA SUSTITUIDOS COMO INHIBIDORES DE ACETIL CoA CARBOXILASA
WO2008088692A2 (en) 2007-01-12 2008-07-24 Merck & Co., Inc. Spirochromanon derivatives
US20100113418A1 (en) 2007-02-20 2010-05-06 Takeda Pharmaceutical Company Limited Heterocyclic compound
EP2110377A1 (en) 2008-04-15 2009-10-21 DAC S.r.l. Spirocyclic derivatives as histone deacetylase inhibitors
JP5435592B2 (ja) 2008-05-28 2014-03-05 ファイザー・インク ピラゾロスピロケトンアセチルCoAカルボキシラーゼ阻害剤
WO2010002010A1 (en) 2008-07-04 2010-01-07 Banyu Pharmaceutical Co.,Ltd. Novel spirochromanone carboxylic acids
EP2318404B1 (en) 2008-07-16 2013-08-21 Merck Sharp & Dohme Corp. Bicyclic heterocycle derivatives and use thereof as gpr119 modulators
EP2318388A2 (en) 2008-07-23 2011-05-11 Schering Corporation Tricyclic spirocycle derivatives and methods of use thereof
AU2009308702B2 (en) 2008-10-31 2015-05-28 Pain Therapeutics, Inc. Filamin A binding anti-inflammatory and analgesic
WO2010051476A1 (en) 2008-10-31 2010-05-06 Pain Therapeutics, Inc. Filamin a-binding anti-inflammatory analgesic
EP2414348B1 (en) 2009-04-03 2013-11-20 Merck Sharp & Dohme Corp. Bicyclic piperidine and piperazine derivatives as gpcr modulators for the treatment of obesity, diabetes and other metabolic disorders
US8354403B2 (en) 2009-08-27 2013-01-15 Merck Sharp & Dohme Corp. Pyrrolidine derived beta 3 adrenergic receptor agonists
CA2812526A1 (en) 2010-05-06 2011-11-10 Vertex Pharmaceuticals Incorporated Heterocyclic chromene-spirocyclic piperidine amides as modulators of ion channels
AU2012212196B2 (en) 2011-02-02 2016-10-13 Vertex Pharmaceuticals Incorporated Pyrrolopyrazine-spirocyclic piperidine amides as modulators of ion channels
US10385070B2 (en) 2011-02-18 2019-08-20 Vertex Pharmaceuticals Incorporated Chroman-spirocyclic piperidine amides as modulators of ion channels

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4353901A (en) * 1981-10-19 1982-10-12 Syntex (U.S.A.) Inc. 9-(1,4-Benzodioxan-2-ylalkyl and hydroxyalkyl)-1-oxa-4,9-diazaspiro[5.5]undecan-3-ones
US5304121A (en) 1990-12-28 1994-04-19 Boston Scientific Corporation Drug delivery system making use of a hydrogel polymer coating
US5886026A (en) 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
US20020013325A1 (en) * 1995-09-29 2002-01-31 Fisher Matthew J. Spiro compounds as inhibitors of fibrinogen-dependent platelet aggregation
US6099562A (en) 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US20060052597A1 (en) * 2002-10-22 2006-03-09 Best Desmond J Aryloxyalkylamine derivatives as h3 receptor ligands
WO2010151595A1 (en) * 2009-06-26 2010-12-29 Schering Corporation Pyrrolo-benzo-1,4-diazines useful as sodium channel blockers
WO2011092198A1 (en) * 2010-01-26 2011-08-04 Boehringer Ingelheim International Gmbh 5-alkynyl-pyrimidines

Non-Patent Citations (43)

* Cited by examiner, † Cited by third party
Title
"Handbook of Chemistry and Physics"
"March's Advanced Organic Chemistry", 2001, JOHN WILEY & SONS
ABRAHAMSEN, B. ET AL.: "The cell and molecular basis of mechanical, cold, and inflammatory pain", SCIENCE, vol. 321, no. 5889, 2008, pages 702
AHMAD, S. ET AL.: "A stop codon mutation in SCN9A causes lack of pain sensation", HUM MOL GENET, vol. 16, no. 17, 2007, pages 2114, XP002517427, DOI: doi:10.1093/HMG/DDM160
AKOPIAN, A. N. ET AL.: "The tetrodotoxin-resistant sodium channel SNS has a specialized function in pain pathways", NAT NEUROSCI, vol. 2, no. 6, 1999, pages 541
AMAYA, F. ET AL.: "The voltage-gated sodium channel Na(v) 1.9 is an effector of peripheral inflammatory pain hypcrscnsitivity", JNEUROSCI, vol. 26, no. 50, 2006, pages 12852
BENNETT, P. B.; YAZAWA, K.; MAKITA, N.; GEORGE, A. L., JR.: "Molecular mechanism for an inherited cardiac arrhythmia", NATURE, vol. 376, no. 6542, 1995, pages 683
CATTERALL, W. A.; GOLDIN, A. L.; WAXMAN, S. G.: "International Union of Pharmacology. XLVII. Nomenclature and structure-function relationships of voltage-gated sodium channels", PHARMACOL REV, vol. 57, no. 4, 2005, pages 3 97, XP055146853, DOI: doi:10.1124/pr.57.4.4
CHAHINE, M.; CHATELIER, A.; BABICH, 0.; KRUPP, J. J.: "CNS Neurol Disord Drug Targets", vol. 7, 2008, article "Voltage-gated sodium channels in neurological disorders", pages: 144
COX, J. J. ET AL.: "An SCN9A channelopathy causes congenital inability to experiencc pain", NATURE, vol. 444, no. 7121, 2006, pages 894, XP009119589, DOI: doi:10.1038/nature05413
CUMMINS, T. R.; SHEETS, P. L.; WAXMAN, S. G.: "The roles of sodium channels in nociception: Implications for mechanisms of pain", PAIN, vol. 131, no. 3, 2007, pages 243, XP022240869, DOI: doi:10.1016/j.pain.2007.07.026
DARBAR, D. ET AL.: "Cardiac sodium channel (SCN5A) variants associated with atrial fibrillation", CIRCULATION, vol. 117, no. 15, 2008, pages 1927
DRENTH, J. P. ET AL.: "SCN9A mutations define primary erythermalgia as a neuropathic disorder of voltage gated sodium channels", JLNVEST DERMATOL, vol. 124, no. 6, 2005, pages 1333
E. W. MARTIN: "Remington's Pharmaceutical Sciences", 1980, MACK PUBLISHING CO.
ENGLAND, S.: "Voltage-gated sodium channels: the search for subtype-selective analgesics", EXPERT OPIN INVESTIG DRUGS, vol. 17, no. 12, 2008, pages 1849
ESTACION, M. ET AL.: "NaV 1.7 gain-of-function mutations as a continuum: A1632E displays physiological changes associated with erythromelalgia and paroxysmal extreme pain disorder mutations and produces symptoms ofboth disorders", JNEUROSCI, vol. 28, no. 43, 2008, pages 11079
FUJIWARA, T.: "Clinical spectrum ofmutations in SCN1A gene: severe myoclonic epilepsy in infancy and related epilepsies", EPILEPSY RES, vol. 70, no. 1, 2006, pages S223
GEORGE, A. L., JR.: "Inherited disorders of voltage-gated sodium channels", J CLIN INVEST, vol. 115, no. 8, 2005, XP055040295, DOI: doi:10.1172/JCI25505
GOLDBERG, Y. P. ET AL.: "Loss-of function mutations in the NaV 1.7 gene underlie congenital indifference to pain in multiple human populations", CLIN GENET, vol. 71, no. 4, 2007, pages 311
GOLDBERG, Y. P. ET AL.: "Loss-of-function mutations in the NaV 1.7 gene underlie congenital indifference to pain in multiple human populations", CLIN GENET, vol. 71, no. 4, 2007, pages 311
HILLE, BERTIL: "Ion Channels of Excitable Membranes", 2001, SINAUER ASSOCIATES, INC.
JERRY MARCH: "Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 1992, JOHN WILEY & SONS, pages: 14 - 16,18-19
KLUGBAUER, N.; LACINOVA, L.; FLOCKERZI, V.; HOFINANN, F.: "Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells", EMBO J, vol. 14, no. 6, 1995, pages 1084, XP002540498
KRAFTE, D. S.; BANNON, A. W.: "Sodium channels and nociception: recent concepts and therapeutic opportunities", CURR OPIN PHARMACOL, vol. 8, no. 1, 2008, pages 50, XP002540499, DOI: doi:10.1016/j.coph.2007.09.007
LAI, J. ET AL.: "Inhibition of neuropathic pain by decreased expression of the tetrodotoxin-resistant sodium channel, NaV1.8", PAIN, vol. 95, no. 1-2, 2002, pages 143, XP002374879, DOI: doi:10.1016/S0304-3959(01)00391-8
LAI, J. ET AL.: "Inhibition of neuropathic pain by decreased expression of the tetrodotoxin-resistant sodium channel, NaVl .8", PAIN, vol. 95, no. 1-2, 2002, pages 143, XP002374879, DOI: doi:10.1016/S0304-3959(01)00391-8
LAIRD, J. M.; SOUSLOVA, V.; WOOD, J. N.; CERVERO, F.: "Deficits in visceral pain and referred hyperalgesia in NaV 1.8 (SNS/PN3)-null mice", J NEUROSCI, vol. 22, no. 19, 2002, pages 8352, XP001191065
LAIRD, J. M.; SOUSLOVA, V.; WOOD, J. N.; CERVERO, F.: "Deficits in visceral pain and referred hyperalgesia in NaV 1.8 (SNS/PN3)-null mice", JNEUROSCI, vol. 22, no. 19, 2002, pages 8352, XP001191065
MARK H. BEERS; ROBERT BERKOW: "The Merck Manual", 1999, MERCK RESEARCH LABORATORIES
MISRA, S. N.; KAHLIG, K. M.; GEORGE, A. L., JR.: "Impaired NaV1.2 function and reduced cell surface expression in benign familial neonatal-infantile seizures", EPILEPSIA, vol. 49, no. 9, 2008, pages 1535
NASSAR, M. A. ET AL.: "Nerve injury induccs robust allodynia and ectopic discharges in NaV 1.3 null mutant mice", MOL PAIN, vol. 2, 2006, pages 33
NASSAR, M. A. ET AL.: "Nociccptor-spccific genc deletion reveals a major role for NaV 1.7 (PN1) in acute and inflammatory pain", PROC NATL ACAD SCI US A, vol. 101, no. 34, 2004, pages 12706
PORRECA, F. ET AL.: "A comparison of the potential role of the tetrodotoxin-insensitive sodium channels, PN3/SNS and NaN/SNS2, in rat models of chronic pain", PROCNATL ACAD SCI USA, vol. 96, no. 14, 1999, pages 7640, XP055019514, DOI: doi:10.1073/pnas.96.14.7640
PRIEST, B. T. ET AL.: "Contribution of the tetrodotoxin-resistant voltage-gated sodium channel NaV 1.9 to sensory transmission and nociceptive behavior", PROC NATL ACAD SCI U S A, vol. 102, no. 26, 2005, pages 9382, XP002374745, DOI: doi:10.1073/pnas.0501549102
S. M. BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
SODERPALM, B.: "Anticonvulsants: aspects of their mechanisms of action", EUR JPAIN, vol. 6, no. A, 2002, pages 3, XP003009563, DOI: doi:10.1053/eujp.2001.0315
THOMAS SORRELL: "Organic Chemistry", 1999, UNIVERSITY SCIENCE BOOKS
TOLEDO-ARAL, J. J. ET AL.: "Identification of PN1, a predominant voltage-dependent sodium channel expressed principally in peripheral neurons", PROC NATL ACAD SCI U S A, vol. 94, no. 4, 1997, pages 1527, XP002454583, DOI: doi:10.1073/pnas.94.4.1527
VICART, S.; STERNBERG, D.; FONTAINE, B.; MEOLA, G.: "Human skeletal muscle sodium channclopathies", NEUROL SCI, vol. 26, no. 4, 2005, pages 194, XP019361583
WANG, G. K.; MITCHELL, J.; WANG, S. Y.: "Block of persistent late Na+ currents by antidepressant sertraline and paroxetine", J MEMBR BIOL, vol. 222, no. 2, 2008, pages 79, XP019622596
WANG, Q. ET AL.: "SCN5A mutations associated with an inherited cardiac arrhythmia, long QT syndrome", CELL, vol. 80, no. 5, 1995, pages 805, XP000919018, DOI: doi:10.1016/0092-8674(95)90359-3
WILLIAMS, B. S. ET AL.: "Characterization of a New Class of Potent Inhibitors of the Voltage-Gated Sodium Channel NaV 1.7", BIOCHEMISTRY, vol. 46, 2007, pages 14693 - 14703, XP009162257, DOI: doi:10.1021/bi7018207
YEOMANS, D. C. ET AL.: "Decrease in inflammatory hyperalgesia by herpes vector-mediated knockdown ofNaV 1.7 sodium channels in primary afferents", ILUM GENE THER, vol. 16, no. 2, 2005, pages 271, XP002517426, DOI: doi:10.1089/hum.2005.16.271

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8598164B2 (en) 2010-05-06 2013-12-03 Vertex Pharmaceuticals Incorporated Heterocyclic chromene-spirocyclic piperidine amides as modulators of ion channels
US8916565B2 (en) 2011-02-02 2014-12-23 Vertex Pharmaceuticals Incorporated Pyrrolopyrazine-spirocyclic piperidine amides as modulators of ion channels
US9511067B2 (en) 2011-02-02 2016-12-06 Vertex Pharmaceuticals Incorporated Substituted spiro[piperidine-4,1'-pyrrolo[1,2-a]pyrazine]s as modulators of ion channels
US10385070B2 (en) 2011-02-18 2019-08-20 Vertex Pharmaceuticals Incorporated Chroman-spirocyclic piperidine amides as modulators of ion channels
US9403839B2 (en) 2012-01-16 2016-08-02 Vertex Pharmaceuticals Incorporated Pyran-spirocyclic piperidine amides as modulators of ion channels
CN104136442A (zh) * 2012-01-16 2014-11-05 沃泰克斯药物股份有限公司 作为离子通道调节剂的吡喃-螺环哌啶酰胺类
WO2013109521A1 (en) * 2012-01-16 2013-07-25 Vertex Pharmaceuticals Incorporated Pyran-spirocyclic piperidine amides as modulators of ion channels
CN104136442B (zh) * 2012-01-16 2016-12-21 沃泰克斯药物股份有限公司 作为离子通道调节剂的吡喃‑螺环哌啶酰胺类
WO2014120815A1 (en) 2013-01-31 2014-08-07 Vertex Pharmaceuticals Incorporated Quinoline and quinazoline amides as modulators of sodium channels
EP3865475A1 (en) 2013-01-31 2021-08-18 Vertex Pharmaceuticals Incorporated Process for preparing pyridone amides which are modulators of sodium channels and intermediate compounds used therein
US11673864B2 (en) 2013-01-31 2023-06-13 Vertex Pharmaceuticals Incorporated Pyridone amides as modulators of sodium channels
WO2014120808A1 (en) 2013-01-31 2014-08-07 Vertex Parmaceuticals Incorporated Pyridone amides as modulators of sodium channels
US11203571B2 (en) 2013-07-19 2021-12-21 Vertex Pharmaceuticals Incorporated Sulfonamides as modulators of sodium channels
WO2015089361A1 (en) 2013-12-13 2015-06-18 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
US11773119B2 (en) 2013-12-13 2023-10-03 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
US9464102B2 (en) 2013-12-13 2016-10-11 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
US9828397B2 (en) 2013-12-13 2017-11-28 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
US10787472B2 (en) 2013-12-13 2020-09-29 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
EP3459958A1 (en) 2013-12-13 2019-03-27 Vertex Pharmaceuticals Incorporated Process and intermediates for preparing prodrugs of pyridone amides useful as modulators of sodium channels
US10253054B2 (en) 2013-12-13 2019-04-09 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
US9163042B2 (en) 2013-12-13 2015-10-20 Vertex Pharmaceuticals Incorporated Prodrugs of pyridone amides useful as modulators of sodium channels
CN106459093A (zh) * 2014-06-02 2017-02-22 埃斯蒂文博士实验室股份有限公司 具有多重模式抗疼痛活性的1‑氧杂‑4,9‑二氮杂螺十一烷化合物的烷基与芳基衍生物
US10065971B2 (en) 2014-06-02 2018-09-04 Esteve Pharmaceuticals S.A. Amide derivatives of 1-oxa-4,9-diazaspiro undecane compounds having multimodal activity against pain
WO2015185207A1 (en) * 2014-06-02 2015-12-10 Laboratorios Del Dr. Esteve, S.A. Alkyl and aryl derivatives of 1-oxa-4,9-diazaspiro undecane compounds having multimodal activity against pain
US10703765B2 (en) 2014-06-02 2020-07-07 Esteve Pharmaceuticals, S.A. Alkyl and aryl derivatives of 1-oxa-4,9-diazaspiro undecane compounds having multimodal activity against pain
US10421750B2 (en) 2015-10-23 2019-09-24 Esteve Pharmaceuticals, S.A. Substituted morpholine derivatives having activity against pain
US10183918B2 (en) 2015-10-23 2019-01-22 Laboratorios Del Dr. Esteve S.A. Oxa-azaspiro compounds having activity against pain
WO2017067665A1 (en) * 2015-10-23 2017-04-27 Laboratorios Del Dr. Esteve, S.A. Substituted morpholine derivatives having activity against pain
WO2017067664A1 (en) * 2015-10-23 2017-04-27 Laboratorios Del Dr. Esteve, S.A. Oxa-diazaspiro compounds having activity against pain
WO2017067663A1 (en) * 2015-10-23 2017-04-27 Laboratorios Del Dr. Esteve, S.A. Oxa-azaspiro compounds having activity against pain
WO2018213426A1 (en) 2017-05-16 2018-11-22 Vertex Pharmaceuticals Incorporated Deuterated pyridone amides and prodrugs thereof as modulators of sodium channels
US11358977B2 (en) 2017-05-16 2022-06-14 Vertex Pharmaceuticals Incorporated Deuterated pyridone amides and prodrugs thereof as modulators of sodium channels
US10647661B2 (en) 2017-07-11 2020-05-12 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
US11603351B2 (en) 2017-07-11 2023-03-14 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
WO2019014352A1 (en) 2017-07-11 2019-01-17 Vertex Pharmaceuticals Incorporated CARBOXAMIDES AS INHIBITORS OF SODIUM CHANNELS
US11529337B2 (en) 2018-02-12 2022-12-20 Vertex Pharmaceuticals Incorporated Method of treating pain
WO2020146682A1 (en) 2019-01-10 2020-07-16 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
WO2020146612A1 (en) 2019-01-10 2020-07-16 Vertex Pharmaceuticals Incorporated Esters and carbamates as modulators of sodium channels
WO2020176763A1 (en) 2019-02-27 2020-09-03 Vertex Pharmaceuticals Incorporated Dosage form comprising prodrug of na 1.8 sodium channel inhibitor
WO2020219867A1 (en) 2019-04-25 2020-10-29 Vertex Pharmaceuticals Incorporated Pyridone amide co-crystal compositions for the treatment of pain
US11834441B2 (en) 2019-12-06 2023-12-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
US11919887B2 (en) 2019-12-06 2024-03-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
WO2021113627A1 (en) 2019-12-06 2021-06-10 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
EP4081509A4 (en) * 2019-12-24 2024-01-03 Zydus Lifesciences Ltd NOVEL COMPOUNDS SUITABLE FOR TREATING DYSLIPIDEMIA
WO2022256702A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran-2-carboxamides as modulators of sodium channels
WO2022256622A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
WO2022256676A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran analogs as modulators of sodium channels
US11827627B2 (en) 2021-06-04 2023-11-28 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
WO2022256679A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamide analogs as modulators of sodium channels
WO2022256842A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Hydroxy and (halo)alkoxy substituted tetrahydrofurans as modulators of sodium channels
WO2022256708A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Solid dosage forms and dosing regimens comprising (2r,3s,4s,5r)-4-[[3-(3,4-difluoro-2-methoxy-phenyl)-4,5-dimethyl-5-(trifluoromethyl) tetrahydrofuran-2-carbonyl]amino]pyridine-2-carboxamide
WO2023205778A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205468A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205465A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205463A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain

Also Published As

Publication number Publication date
US9181273B2 (en) 2015-11-10
JP2014508169A (ja) 2014-04-03
MX347982B (es) 2017-05-22
AU2012229187A1 (en) 2013-09-19
MX2013010476A (es) 2013-10-30
JP6088443B2 (ja) 2017-03-01
CA2829803A1 (en) 2012-09-20
US20150025073A1 (en) 2015-01-22
AU2012229187B2 (en) 2016-11-10
US8828996B2 (en) 2014-09-09
CN103517910B (zh) 2016-12-14
CN103517910A (zh) 2014-01-15
EP2686325A1 (en) 2014-01-22
NZ615014A (en) 2016-08-26
ES2618929T3 (es) 2017-06-22
EP2686325B1 (en) 2016-12-14
US20120264749A1 (en) 2012-10-18

Similar Documents

Publication Publication Date Title
AU2012229187B2 (en) Morpholine-spirocyclic piperidine amides as modulators of ion channels
US9511067B2 (en) Substituted spiro[piperidine-4,1'-pyrrolo[1,2-a]pyrazine]s as modulators of ion channels
AU2013209958B2 (en) Pyran-spirocyclic piperidine amides as modulators of ion channels
AU2012217616B2 (en) Chroman - spirocyclic piperidine amides as modulators of ion channels
US8598164B2 (en) Heterocyclic chromene-spirocyclic piperidine amides as modulators of ion channels
AU2014287471C1 (en) Fused piperidine amides as modulators of ion channels
WO2014022639A1 (en) Solid forms of (4-isopropoxy-3-methoxyphenyl)(2'-methyl-6'-(trifluoromethyl)-3',4'-dihydro-2'h-spiro[piperidine-4,1'-pyrrolo[1,2-a]pyrazine]-1-yl)methanone
NZ615014B2 (en) Morpholine-spirocyclic piperidine amides as modulators of ion channels

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12710841

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2829803

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013558104

Country of ref document: JP

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2012710841

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/010476

Country of ref document: MX

Ref document number: 2012710841

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2012229187

Country of ref document: AU

Date of ref document: 20120313

Kind code of ref document: A